2017 PHYS REV Grelina CCK glp1 Pyy Papeis Fisiológicos Na Obesidade Saúde PDF
2017 PHYS REV Grelina CCK glp1 Pyy Papeis Fisiológicos Na Obesidade Saúde PDF
2017 PHYS REV Grelina CCK glp1 Pyy Papeis Fisiológicos Na Obesidade Saúde PDF
University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia
Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM
Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology,
University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University
Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University,
New York, New York
L
Ghrelin, CCK, GLP-1, and PYY(3–36): Secretory Controls and Physiological Roles in
Eating and Glycemia in Health, Obesity, and After RYGB. Physiol Rev 97: 411– 463,
2017. Published December 21, 2016; doi:10.1152/physrev.00031.2014.—The
efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the man-
agement of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI)
endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-
related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the
secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-
1), and peptide tyrosine tyrosine [PYY(3–36)], and their contributions to the controls of GI motor
function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons,
as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting
signals are discussed. Gastric emptying, the detection of specific digestive products by small
intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute
to the secretion of ghrelin, CCK, GLP-1, and PYY(3–36). While CCK has been fully established as
an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones
in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contrib-
utes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in
these hormones’ actions, but methods to determine the physiological status of local signaling
effects are lacking. Further research and fresh approaches are required to better understand
ghrelin, CCK, GLP-1, and PYY(3–36) physiology; their roles in obesity and bariatric surgery; and
their therapeutic potentials.
meals and were not thought to be relevant for the tonic ingly important issue in GI endocrinology, whether the hor-
control of total energy intake and body-weight regulation mones’ mode of signaling in these situations is classically
(e.g., Refs. 172, 521, 672, 837). This view soon changed. In endocrine or local; 4) given the close relationship of GI
2002, Cummings et al. (174) reported that levels of the endocrine and GI motor physiology, the role of GI motility
gastric hormone ghrelin, which had been shown to increase in the hormones’ effects; 5) whether obesity [i.e., body mass
eating when infused intravenously in humans (839), were index (BMI); weight in kg/(height in m)2 ⱖ30 kg/m2], alters
inversely related to body adiposity in healthy-weight, obese, the hormones’ effects on eating or glycemic control, and 6)
and weight-reduced humans, consistent with a tonic signal- because of the marked alterations in nutrient delivery into
ing function. Recent clinical trials indicate that treatment the small intestines and contact with enteroendocrine cells
with long-acting glucagon-like peptide-1 (GLP-1) receptor after RYGB (FIGURE 1), the hormones’ contributions to the
agonists such as liraglutide [Victoza for type 2 diabetes effects of RYGB on eating and glycemic control.
mellitus (T2DM) and Saxenda for weight control, Novo
Nordisk, Bagsvaerd, Denmark] leads to weight loss and
amelioration of T2DM (335, 414, 581). Finally, changes in B. Approach
GI hormone secretion provide plausible mechanisms for the
remarkable therapeutic efficacy of bariatric surgery, espe- 1. Why focus on meals?
cially Roux-en-Y gastric bypass (RYGB), to reduce adipos-
ity and improve glycemic control (113, 310, 421, 513, 551, Total amount eaten and glycemic control are critically de-
712). pendent on the control of and physiological responses to
individual meals, and a significant component of these func-
In light of this, we review, 1) the secretion of ghrelin, CCK, tions is thought to be mediated by ghrelin, CCK, GLP-1,
GLP-1, and peptide tyrosine tyrosine [PYY(3–36)] around and PYY(3–36) secretion, as schematized in FIGURE 2.
meals; 2) the contributions of these hormones to the control
of meal size, meal timing, and meal-related glycemia, but as The timing, size, and content of meals provide a complete
explained below, not to hedonics; 3) because it is an increas- description of what, when, and how much (in terms of g,
A B
Ghrelin CCK GLP-1 PYY Intact GI tract GI tract after RYGB
Stomach
Small intestine
Colon
FIGURE 1. Schematic depictions of the localization of enteroendocrine cells and changes after RYGB.
A: distribution of enteroendocrine cells secreting ghrelin, CCK, GLP-1, and PYY in the stomach (pink),
duodenum (yellow), jejunum (green), and ileum (violet). Black areas indicate the relative densities of expression
of enteroendocrine cells producing the hormones indicated. Enteroendocrine cells secreting particular hor-
mones were initially categorized histologically, e.g., I cells for CCK, L cells for enteroglucagons and PYY, etc.
(166, 567, 591). It is now clear, however, that this categorization is not a reliable guide to hormone secretion.
Rather, individual enteroendocrine cells secrete variable mixtures of hormones (231, 303, 597, 738).
Bottom salmon rectangle, proximal large intestine. B: intact gastrointestinal tract (left) and gastrointestinal
rearrangement after RYGB (right). Pink areas are stomach, salmon areas are large intestine (⬃1.5 m long in
healthy adults), yellow is duodenum (typically ⬃25 cm long), green is jejunum (⬃2–3 m), and violet is ileum
(⬃3– 4 m). For RYGB, the stomach is divided into a small upper pouch with a volume of ⬃25 ml and an isolated
gastric remnant, the small intestine is divided ⬃50 cm from the pylorus, and the distal limb of the small
intestine (Roux or alimentary limb) is brought up to the gastric pouch and connected to it by an end-to-side
gastroenterostomy. As a result, ingested food enters the small gastric pouch and empties directly into the
jejunum. The gastric remnant and isolated ⬃50 cm of small intestine (“biliopancreatic limb”) is connected
to the jejunum ⬃150 cm distal to the gastroenterostomy. The small intestine distal to the anastomosis is
called the common channel.
kcal, or macronutrients) is eaten. Meal patterns are pro- shift from nutritional homeostasis to behavioral neurosci-
duced by species-specific physiological controls as well as ence.”
environmental, social, and cultural contingencies. The con-
trol of meal size is disturbed in psychiatric eating disorders Ghrelin, CCK, GLP-1, and PYY(3–36) contribute to three
(271, 483, 743). In addition, obese individuals eat larger of the putative motivational processes that provide the basic
meals than healthy-weight individuals (5, 74, 189, 497) unconditioned control of meal initiation and meal size (85,
(healthy body weight is BMI ⱖ18.5 and ⬍25 kg/m2). Thus 86, 271): 1) hunger, which refers to the process energizing
the physiology of individual meals is crucial for understand- the acquisition of food and meal initiation; 2) satiation,
ing normal and disordered eating, including the chronic which leads to ending the meal; and 3) postprandial satiety,
overeating that has led to the obesity epidemic (368, 741, which inhibits eating after meals and prolongs the intermeal
792). Smith (702) referred to the recognition of the central interval. The hormones’ possible roles in a fourth meal-
role of meals in the physiology of eating as “a paradigm control process, flavor hedonics, and the central neural
mechanisms integrating their effects are not reviewed, as
these topics have been adequately reviewed elsewhere (for
reviews of the hormones’ hedonic effects, see Refs. 24, 212,
467, 499, 571, 699, 700, 827; for reviews of their central
processing, see Refs. 75, 76, 291, 524, 623, 625).
The ability to analyze hormone function with agonists and onto gastric emptying. Therefore, the review begins with an
antagonists (criterion 6) is linked to developments in recep- introduction to the effects of GI motor function on eating
tor pharmacology and receptor-subtype analyses. The use and glycemia in health, obesity, and after RYGB.
of antagonists in particular is now considered one of the
cardinal criteria for physiological function. These tools also
II. GI MOTOR FUNCTION
demand careful interpretation if the biological half-life, re-
ceptor affinity, relative access to receptors beyond the
blood-brain barrier, etc., differ between the hormone and A. Gastric Accommodation and Emptying
the agonist or antagonist. For example, the eating-inhibi-
tory effects of the long-lasting GLP-1 agonist exendin-4 Physical digestion of solid food begins in the mouth, but is
differ markedly from those of native GLP-1. primarily a gastric function (126, 127, 318, 341, 366, 481,
578, 724). Gastric volume during the meal usually exceeds
3. Why consider GI motor function? the volume of ingesta due to gastric secretions and swal-
lowed saliva and air (118, 282). Vago-vagal gastric-accom-
GI endocrine function and GI motor function are so closely modation reflexes increase gastric volume as meals prog-
related that one cannot be understood without the other. ress, avoiding significant increases in intragastric pressure
Gastric emptying and intestinal transit determine which en- or gastric-wall tension (43, 405). The lack of stimulation of
teroendocrine cells are exposed to chyme and for how long. gastric-tension receptors ensures that accommodation does
This in turn affects GI hormone secretion, which feeds back not lead to aversive sensations, although they do appear
sufficient to elicit a pleasant sensation of fullness (215, 464).
Accommodation reflexes are triggered mainly by gastric
IE LMM
mechanoreceptors and intestinal nutrient receptors and are
mediated in part by CCK (43, 246).
Lumen Lamina
propria
FIGURE 3. Schematic of the small intestinal mucosa showing po-
tential modes of action of CCK, GLP-1, and PYY. The mucosa in-
cludes the epithelial cell layer (IE) on the luminal side, the lamina
propria, and the lamina muscularis mucosae (LMM), which limns
the submucosa and underlying serosa (not shown). The epithelium
consists of enterocytes (tan), which are specialized for nutrient
1 Endocrine absorption, enteroendocrine cells (blue, villi not shown), which se-
Digested crete GI hormones, and other cell types (not shown). Digested nu-
nutrients trients activate specific nutrient receptors and transporters (orange
⬍) expressed on the apical surface of enteroendocrine cells, leading
to secretion of CCK, GLP-1, and PYY from the basolateral side of
2 Neurocrine enteroendocrine cells. Four modes of action are diagrammed. Mode
1 is the classical endocrine mode, in which hormones diffuse from
the lamina propria into mesenteric capillaries (salmon), which drain
into the hepatic-portal vein and finally the systemic circulation, allow-
ing hormones to act on distant targets. Modes 2– 4 show variations
3 Paracrine of local actions. Mode 2 is a neuroendocrine mode, in which hor-
mones in the lamina propria activate vagal afferents (green arrow),
which in turn stimulate brain-mediated responses. Mode 3 is the
paracrine mode, in which hormones in the lamina propria act on
receptors (black ⬍) on nearby cells, either neuroendocrine cells or
other cell types. Mode 4 shows the anatomical basis for a neuropod
mode of action, which has been described for enteroendocrine CCK
4 Neuropod and PYY cells, and may exist for other GI hormones. This involves
hormone release from enteroendocrine-cell neuropods that end in
synapse-like appositions to glial cells of the enteric nervous system
and other cell types. Note that the hormone concentrations involved
Afferent in these different modes vary: hormone concentrations in the small
gap between neuropods and adjacent cells are likely to be highest,
paracrine and vagal neuroendocrine signaling may be the next high-
est hormone concentrations, endocrine signaling in the liver involves
moderate hormone concentrations, and endocrine signaling in
which hormones reach their receptors via the systemic circulation
involves relatively low hormone concentrations. Hormones also en-
ter the lymph from the lamina propria via bulk flow (not shown), but
Efferent this is not known to be functionally relevant. Although ghrelin secre-
tion is not stimulated directly by nutrients, secreted ghrelin may act
in the modes shown here.
100 0
Table 3. Physiological endocrine doses of ghrelin, CCK, GLP-1,
and PYY(3–36) in healthy-weight humans 80 20
Solid
food
% Remaining
% Emptied
Physiological Dose,
Hormone pmol·kgⴚ1·minⴚ1 Reference Nos. 60 40
contrast, intraindividual variability is low under laboratory tying elicited by intragastric lipid infusion (374), but
conditions (532). whether reproducing endogenous amounts and patterns of
ghrelin is sufficient to stimulate gastric emptying and
Neural and endocrine reflexes are generally thought to syn- whether ghrelin antagonists inhibit gastric emptying have
ergize in the control of gastric emptying (126, 216, 555). not been tested in humans. Thus ghrelin does not yet fulfill
The importance of the vagal contribution is underscored by criteria 3 and 6 (TABLE 2) for having an endocrine role in
the decrease in emptying of solid foods after vagotomy gastric emptying. Similarly, a supraphysiological ghrelin in-
(405). The roles of ghrelin, CCK, GLP-1, and PYY(3–36) fusion elicited phase III MMC activity, but smaller doses
are reviewed in section IIC. did not (203, 748), and endogenous phase III MMC activity
was not temporally associated with plasma ghrelin concen-
trations (although phase III MMC activity was associated
B. Small Intestinal Motility with motilin levels) (204). Thus these tests did not produce
evidence that ghrelin fulfills criteria 1 or 3 (TABLE 2) for an
The contributions of segmentation, mixing, and propulsion endocrine effect on GI motility.
of chyme in the small intestine to intestinal nutrient sensing
and the control of eating and glycemia are not well under- Several studies indicate that CCK meets both criteria 1 and
stood. Challenges include: 1) present methods to measure 6 (TABLE 2) for having an endocrine role in gastric emptying
segmentation, mixing, and propulsion of chyme are limited, of liquid food (262, 263, 436, 447, 495, 674; but see Ref.
although novel approaches may soon accelerate progress 433 for a negative report). Animal studies indicate that
(30, 33, 87, 220); and 2) the simultaneous changes in gastric CCK slows gastric emptying via vago-vagal reflexes stimu-
accommodation, gastric emptying, and GI-hormone secre-
lated by both endocrine and paracrine signaling (216, 555,
tion are difficult to control (641, 681, 682, 718). Multivar-
823). CCK also fulfilled criteria 1, 4, and 6 for endocrine
iate statistics, such as the approaches of Seimon et al. (683)
roles in the increases in tonic and phasic pyloric pressures
and Acosta et al. (5), provide a useful strategy to dissect
and reductions in antral and duodenal pressures stimulated
these diverse factors functionally.
by intraduodenal lipids (102, 190, 259, 316, 584), re-
sponses that presumably contribute to CCK’s inhibitory
Two pharmacological studies suggest an important role for
effect on gastric emptying (382). One study in humans
small intestinal motility in incretin hormone secretion. In
failed to detect an effect of CCK-receptor antagonism on
both, healthy-weight subjects received intraduodenal infu-
small intestinal transit time (495).
sions of glucose, and intraduodenal pressure and flow
events were assessed by manometry and impedance mea-
surements. Pretreatment with hyoscine butylbromide (137) There is also support for GLP-1 as an endocrine control of
increased intraduodenal waves for 10 min and reduced flow gastric emptying. Physiological doses of GLP-1 slowed
events for 60 min. This was associated with decreased emptying of liquid meals (541, 822), supporting criterion 3
(TABLE 2), and administration of the GLP-1 receptor antag-
plasma level GIP at 10 and 20 min, suggesting that normal
GIP release depends on the spread of glucose through duo- onist exendin(9 –39) accelerated gastric emptying in two
denum and proximal jejunum. In contrast, pretreatment studies (41, 196), supporting criterion 6. Exendin(9 –39)
with metoclopramide (411) stimulated duodenal pressure failed to affect gastric emptying in three other studies (531,
waves, but did not affect flow events. Metoclopramide pro- 546, 650), however, suggesting that differences in test meal
duced marked increases in plasma GLP-1 and GIP, suggest- characteristics, plasma glucose levels, or other situational
ing that increased mixing of the luminal contents increased variable may contribute to GLP-1’s influence on gastric
their contact with enteroendocrine cells, thus increasing in- emptying. Exendin(9 –39) also stimulates PYY(3–36) secre-
cretin secretion. By extension, small intestinal motility may tion, which may slow gastric emptying and contribute to
also affect ghrelin, CCK, and PYY secretion. these variable results (41, 230, 665, 670, 721, 843). Intra-
venous infusion of physiological doses of GLP-1 also stim-
When the stomach is empty, small intestinal peristaltic ac- ulated tonic and phasic pyloric pressures and reduced antral
tivity is controlled by phase III activity of the migrating and duodenal pressure waves (662), and infusion of exen-
motor complex (MMC), which originates in the stomach in din(9 –39) abolished glucose-induced changes in antropylo-
humans, rats, and mice (203, 266, 748, 853) and appears to roduodenal pressures (664), indicating a role for GLP-1 in
be stimulated by motilin (203). small intestinal motility.
GLP-1 infusions (199) each decreased eating more than the 497) in obese relative to healthy-weight people. These in-
individual manipulations. consistent results may be related to several differences
among the studies, including differences in the nutrient
Gastric-volume signals are thought to control eating via composition of the test meals and methodological differ-
mechanoreceptors that are tuned to both tension and ences (e.g., scintigraphy vs. less direct measures). In con-
stretch or length (294, 366, 560, 579) and are linked to the trast, a large scintigraphic study (389 subjects) demon-
CNS by vagal and spinal visceral (splanchnic) afferents strated clearly that overweight and obesity are associated
(272, 791, 809). In rats, these include polymodal vagal with increased gastric emptying rates of both solid and liq-
afferents whose response can be increased severalfold by uid foods (5). Interestingly, the degrees of increase were
combinations of gastric fill and CCK infusion (671) or in- similar in overweight, obese, and morbidly obese (i.e., BMI
fluenced oppositely by CCK and ghrelin treatment (217). ⱖ35 kg/m2) subjects (decreases of ⬃20% in solid-meal half-
Thus neural information processing controlling GI function emptying time and ⬃30% in liquid-meal half-emptying
and eating appears to begin at the level of the vagal affer- time in each group). This suggests increased gastric-empty-
ents. ing rate is more likely to be a permissive rather than an
effective cause of obesity.
As mentioned above, negative-feedback loops link gastric
volume and gastric emptying to intestinal nutrient sensing Manipulations of gastric volume may contribute to obesity
and to ghrelin, CCK, GLP-1, and PYY(3–36) secretion (FIG- therapy. Consistent with this possibility, some data relate
URE 5). The relationship of these feedback loops to satiation is the eating-inhibitory, weight-reducing, and glycemic effects
complex because both gastric and postgastric signals contrib- of the GLP-1 receptor agonist liraglutide to reduced gastric
ute to satiation. Thus, depending on the circumstances, accel- emptying (343, 789). Furthermore, implantable devices de-
erating gastric emptying may either decrease satiation by de- signed to electrically stimulate the vagus in a way that
creasing gastric-volume signals or increase satiation by in- blocks vagal signaling reduced subjective hunger, increased
creasing postgastric signals. This point is underscored by fullness, decreased body weight, and improved glycemic
the report (770) that erythromycin accelerated gastric emp- regulation in clinical trials (89, 134, 353, 654, 656, 689).
tying and decreased rather than increased meal size in a The most compelling of these was a randomized, double-
group of overweight (i.e., BMI ⱖ25 and ⬍30 m/kg2) and blind, sham-controlled trial involving 239 obese patients
obese (BMI ⱖ30 kg/m2) subjects. Finally, recent data sug- (353). Those receiving vagal blockade lost 24% of their
gest functional roles for gastric nutrient-sensing receptors, excess weight in 1 year, versus 16% in the sham-operated
which may include roles in eating (see sect. IIIC). group. The mechanism underlying the efficacy of vagal
blockade is unknown. One possibility is that slowed gastric-
E. Glycemic Control emptying rate is involved (134, 405). In the patients de-
scribed above, gastric emptying was reportedly unchanged,
Gastric emptying contributes importantly to the regulation but because gastric emptying was measured after the pa-
of meal-related glycemia and, thus, to overall glucose ho- tients had undergone more than a year of vagal stimulation,
meostasis (472). For example, in both healthy subjects and it is possible that there was tachyphylaxis of an earlier effect
patients with T2DM, intersubject variability in emptying of (658). In other studies, vagal blockade increased ghrelin
an oral glucose load accounted for significant amounts of secretion and reduced secretion of CCK and GLP-1 (153,
the variability in plasma glucose increments (342, 470). In 154), effects that presumably would oppose any decrease in
addition, pharmacological manipulation of gastric empty- eating. An alternative hypothesis that deserves investigation
ing of a solid-liquid mixed-nutrient meal produced corre- is that vagal blockade reduces gastric accommodation dur-
sponding glycemic changes in patients with T2DM (285). ing meals, leading to increased distension and early satia-
Variation in factors that affect gastric emptying, such as tion. Because the blockade prevents vagal afferent signal-
decreasing dietary fiber content (369, 771), would presum- ing, this hypothesis requires that distension is adequately
ably increase the relative contribution of gastric emptying sensed by spinal-visceral afferents (82, 146).
to meal-related glycemia; conversely, manipulating post-
gastric factors, such as incretin-hormone secretion, would
reduce it. Small intestinal nutrient transport also may affect G. RYGB
meal-related glycemia. For example, pharmacological slow-
ing of intestinal flow of intraduodenally infused glucose Due to a greatly reduced gastric lumen (FIGURE 1), only a
slowed glucose absorption and reduced blood glucose fraction of the normal gastric volume can be accommo-
(137). dated, and antral trituration and pyloric control of empty-
ing are absent after RYGB. As a result, RYGB markedly
F. Obesity accelerates gastric emptying of liquids and solids (although
emptying of small, solid meals with volumes not exceeding
In several small-scale studies, gastric emptying was compa- the pouch volume may be slower) (213, 244, 339, 518, 536,
rable (562, 681), faster (295, 794, 797), or slower (338, 802, 808). This, in turn, often leads to bloating, nausea, and
dumping in RYGB patients (307, 377, 545, 695, 745). Glu- amus (Arc) (184, 398, 524, 597, 742, 799, 826). Ghrelin
cose solutions, as used in glucose-tolerance tests, may O-acyltransferase (GOAT) catalyzes the conversion of
empty almost immediately in RYGB patients, leading to the ghrelin into its biologically active acylated forms, octanoyl-
appearance of ⬃300 kcal in the jejunum within 1–2 min and decanoyl-ghrelin (together referred to as acyl-ghrelin,
(544). Such rapid increases in small intestinal nutrient con- in contrast to unacylated or des-acyl-ghrelin). GOAT phys-
tent are likely to contribute to the increased meal-related iology has emerged as an important modulator of ghrelin
secretion of CCK, GLP-1, PYY(3–36), and insulin after function (58, 389, 524, 847). Less than 10% of circulating
RYGB, as described in the next sections. For example, in- ghrelin is acyl-ghrelin, which together with the difficulty in
fusion of glucose at a high physiological rate (4 kcal/min) assaying it, complicates studies of endogenous ghrelin (390,
into the Roux limb of RYGB patients and into the duode- 596, 707). The ghrelin receptor was described in 1996 as
num of healthy subjects elicited comparable increases in the growth hormone-secretagogue receptor-1A (GHSR1A)
GLP-1, whereas oral glucose loads (200 kcal/150 ml) pro- (348). It is widely expressed peripherally and centrally (398,
duced larger GLP-1 responses in the RYGB patients (544). 524, 799). Des-acyl-ghrelin has little affinity for GHSR1A
but may have metabolic effects via other receptors (524).
Three additional studies reveal further contributions of
RYGB-induced alterations of GI function to changes in eat-
ing and body weight: 1) faster pouch emptying on postop- B. Secretion
erative day 1 was associated with a ⬃4 kg increase in 1 year
weight loss (21); 2) pouch size was negatively correlated Plasma concentrations of total and acyl-ghrelin increase
with weight loss after 6 months and 1 year (626); and 3) before meals, decline precipitously after meals, and then
thresholds for detection of inflation of a balloon placed in increase gradually until the next meal (173, 326, 707). For
the Roux limb were negatively correlated with spontaneous example, when acyl-ghrelin was sampled frequently
meal sizes 6 months and 1 year postoperatively (81). Re- throughout the day in subjects adhering to a controlled
learning to eat comfortably is likely to be important in some sleep-wake, activity and meal protocol (707), acyl-ghrelin
of these effects, but such learning has not yet been studied maxima were ⬃110 pM before breakfast and ⬃100 pM
much in either humans (108, 177) or animals (424, 690). before lunch and dinner, and post-meal minima were ⬃70
For example, a questionnaire follow-up indicated that meat pM. Importantly, the ratio of circulating acyl- to total ghre-
was the food most often linked to food aversions after lin may change around meals (707). Because ghrelin’s
RYGB (288), which may be due to the challenge of digest- plasma half-life is ⬃30 min (20, 800), postprandial acyl-
ing meat without a stomach. ghrelin dynamics presumably primarily reflect inhibition of
secretion. There is also a gradual decrease in acyl-ghrelin
after midnight, which probably reflects an inhibitory effect
H. Summary of sleep; the pre-breakfast increase begins only after awak-
ening (707). These patterns suggest that habitual sleep-
GI motor function and gastric emptying are closely regu- wake cycles and the timing of breakfast modulate morning
lated. Neural gastric-volume detection contributes to the ghrelin levels. This complicates any definition of “basal”
inhibitory control of eating, and gastric emptying contrib- plasma ghrelin and indicates that across-group compari-
utes to meal-related glycemic control. CCK, GLP-1, and sons of pre-meal plasma ghrelin concentrations should con-
PYY(3–36) contribute to the control gastric emptying, and sider the times of sampling with respect to habitual meal
ghrelin may do so. Intestinal-nutrient sensing links the se- times. Average daily ghrelin levels might provide a useful
cretion of ghrelin, CCK, GLP-1, and PYY(3–36) to gastric alternative. Cummings et al. (173) reported 1) a correlation
emptying and gastric volume (FIGURE 5). Loss of normal of 0.95 between the 0930 h ghrelin level (i.e., the post-
gastric accommodation, food storage, food trituration, and breakfast minimum) and the 24 h ghrelin area under the
emptying are likely to play important roles in the effects of curve (AUC), and 2) a correlation of 0.87 between the 0600
RYGB on eating and glycemic control. h ghrelin level (the pre-breakfast minimum) and the 24 h
AUC, indicating that both measures accurately reflect the
III. GHRELIN integrated or average daily ghrelin level.
A Ghrelin cells are closed-type B CCK, GLP-1 and PYY cells are open-type
Gastric Small-intestinal
1
lumen lumen
3 2
1 4
2
4 3
FIGURE 6. Schematic of the organization of ghrelin, CCK, GLP-1, and PYY entroendocrine cells. A: gastric
ghrelin cells (blue) are closed-type. Their apical aspects are enclosed by epithelial cells (tan) so that they have
no direct contact with the gastric lumen. 1) Neural signals provide the major stimulatory control of ghrelin
secretion. 2) Secreted ghrelin (red dots) diffuses through the lamina propria (yellow) into gastric capillaries
(salmon) and is transported into the hepatic-portal vein and systemic circulation. 3) Ghrelin cells express a
number of nutrient receptors, mainly on the basal and lateral aspects (orange ⬍). These are probably
stimulated mainly by metabolites reaching them by diffusion from the gastric capillaries through the lamina
propria, although some nutrients may reach them directly from the stomach. 4) CCK, PYY(3–36), perhaps
other small intestinal hormones, and other humoral stimuli reach ghrelin cells via the circulation and inhibit
ghrelin secretion. Paracrine signals (not shown) may also be involved. B: CCK, GLP-1, and PYY cells (blue) are
open-type, with direct contact with the small intestinal lumen. 1) Each expresses a number of nutrient
receptors, mainly on the apical and lateral aspects (orange ⬍). These are probably the major controls of
secretion of these hormones. The nutrient receptors expressed by ghrelin, CCK, GLP-1, and PYY cells are
listed in TABLE 4, which also indicates the extensive overlap in the nutrient receptors expressed by the these
cell types. 2) Secreted hormones (red dots) diffuse through the lamina propria (yellow) into small-intestinal
capillaries (salmon) and are transported into the hepatic-portal vein and systemic circulation. 3) Metabolites,
hormones, and other humoral factors reach CCK, GLP-1, and PYY cells by diffusion from mesenteric capillaries
through the lamina propria (yellow) or from nearby small-intestinal epithelial cells (tan). 4) Neural inputs also
control CCK, GLP-1, and PYY secretion.
512, 696) and inhibit ghrelin secretion (35) in humans. probably stimulated mainly by metabolites entering the
Time cues also increase pre-meal ghrelin levels in sched- laminal propria from the circulation, as discussed below.
ule-fed rats (180, 494). There is some evidence, however, that they can be stimu-
lated by gastric contents. Lu et al. (457), for example, found
Ghrelin secretion after meals is inhibited by GI signals that that mouse ghrelin cells express the free fatty acid receptor
are recruited rapidly by nutrient ingestion. Conditioned 4 (Ffar4), that fatty acids inhibited ghrelin secretion in
(167) and cephalic-phase reflexes (315) may contribute. vitro, and that intragastric lipid loads reduced serum ghre-
There appears to be no gastric phase to post-meal ghrelin lin levels in mice with ligated pylori. Similar results were
inhibition because 1) intragastric water or liquid-diet infu- obtained in rat gastric explants (22, 692). These data are
sions had no effect on ghrelin levels in rats when infusates inconsistent with the rat pyloric cuff data described above
were confined to the stomach with a pyloric cuff (558, 829), (558, 829), and relevant studies remain to be done in hu-
2) plasma ghrelin concentrations were reduced comparably mans. Few human enteroendocrine ghrelin cells express
by intragastric and intraduodenal glucose infusions in GNAT3, TAS1R1/TAS1R3, or FFAR4, although nearby
healthy-weight men and women (563, 719), and 3) in con- cells do, suggesting the possibility of a gastric paracrine
trast to most enteroendocrine cells, gastric ghrelin cells are chemosensory control of ghrelin secretion (825).
closed type, i.e., do not directly contact to the GI lumen
(FIGURE 6A). Nevertheless, gastric ghrelin cells express sev- The intestinal phase of post-meal ghrelin inhibition is well
eral nutrient-sensing receptors that may affect ghrelin secre- established (169, 248, 563, 640, 719). The critical site for
tion (207, 237, 311, 367, 457, 524, 824, 825) (TABLE 4, inhibition by glucose appears to be distal to the duodenum
which includes the full and the former names of the nutrient and proximal jejunum because ghrelin secretion (60 min
receptors discussed below). Because these are expressed AUC) was not inhibited by intraduodenal infusions of glu-
mainly on the basolateral aspects of ghrelin cells, they are cose that were limited to only the proximal 60 cm of the
Table 4. Nutrient receptors expressed by enteroendocrine ghrelin, CCK, GLP-1, and PYY cells
Nutrient Receptor Ghrelin CCK GLP-1 PYY
CASR (CaR) X X X X
CD36 X
FFAR1 (GPR40) X
FFAR2 (GPR43) X X
FFAR3 (GPR41) X
FFAR4 (GPR120) X X X
GNAT3 (gustducin) X X
GPR119 X
HCAR1 (GPR81) X
LPAR5 (GPR93) X
SLC2A1 (GLUT1) X
SLC2A2 (GLUT2) X
SLC2A5 (GLUT5) X
SLC5A1 (SGLT1) X
SLC15A1 (PEPT1) X X
TAS1R1/TAS1R3 (T1R1/T1R3) X X
TAS1R2/TAS1R3 (T1R2/T1R3) X X
TAS1R3 (T1R3) X
The table is based on the evidence of receptor expression in mice, rats, or humans discussed in the text.
Former names of the receptors are given in parentheses. CaR, calcium receptor; CASR, calcium-sensing
receptor; CD36, thrombospondin receptor; FFAR, free fatty acid receptor; GLUT, glucose transporter;
GNAT3, guanine nucleotide-binding protein, alpha transducing 3; GPR, G protein-coupled receptor; HCAR1,
hydroxycarboxylic acid receptor 1; LPAR5, lysophosphatidic acid receptor 5; PEPT1 and SLC15A1, solute
carrier family 15 (oligopeptide transporter), member 1; TAS1R1 and T1R1, taste receptor, member 1;
TAS1R2 and T1R2, taste receptor, member 2; TAS1R3 and T1R3, taste receptor, member 3 (T1R1/T1R3).
Note that abbreviations are for the human genes, although many of the receptors indicated have been
identified on the respective enteroendocrine cells so far only in mice or rats.
small intestine to glucose by an inflated balloon, but was All three macronutrients inhibit ghrelin secretion after
inhibited when glucose was also allowed access to the more meals. Consumption of carbohydrate and protein reduced
distal small intestine (445). The underlying mechanisms are ghrelin levels during the next 3 h more than did isoenergetic
unknown. lipid loads (258, 510). Whether carbohydrates and proteins
differentially affect ghrelin secretion is less clear. 1) In over-
Circulating metabolites and hormones may also contribute weight and obese men, ⬃250 kcal oral loads containing
to the inhibition of ghrelin secretion. Intravenous glucose 80% energy as lactose, whey or casein reduced ghrelin lev-
infusion, alone or together with insulin, reduced ghrelin els more than similar glucose loads 120 –180 min after in-
levels under several conditions (254, 506, 526, 644, 688). gestion (96). 2) In healthy-weight and overweight men and
Insulin may be the key factor, as meals did not reduce ghre- women, ⬃500 kcal oral protein and glucose loads reduced
lin levels in patients with type 1 diabetes mellitus (T1DM) ghrelin levels similarly for ⬃3 h, but protein reduced ghrelin
in the absence of insulin therapy, but did so following rein- levels more effectively subsequently (258). 3) In healthy-
statement of basal euinsulinemia (526). In contrast to glu- weight women, no differences in ghrelin levels were de-
cose infusions, intravenous lipid infusions failed to affect tected during 24 h trials comparing a 10% protein-energy
plasma ghrelin levels (506). Increases in peripheral concen- diet, a 60% carbohydrate-energy diet, and a 30% protein-
trations of lactate and short-chain fatty acids resulting from and 40% carbohydrate-energy diet (427). Carbohydrate
colonic fermentation of poorly digestible carbohydrates type is also important: oral glucose reduced ghrelin levels
(46, 523, 753, 758) may be sensed by hydrocarboxylic acid less than lactose (96), but more than fructose (755). Because
receptor 1 (HCAR1) and FFAR2, respectively, because the none of the studies reviewed above assessed gastric empty-
corresponding receptors are expressed by gastric ghrelin ing, differential rates of small intestinal appearance of in-
cells in mice (237). Plasma lactate also increases following gested nutrients may have contributed as well as direct ef-
many meals (694, 734) as well as during exercise and hyp- fects of specific intestinal nutrient sensors.
oxia (135, 286, 815), and both exercise and hypoxia de-
crease plasma ghrelin levels in rats and humans (135, 815). Lipids and di- or polysaccharides require digestion to in-
Finally, circulating amino acids may inhibit ghrelin secre- hibit ghrelin secretion fully because tetrahydrolipstatin, a
tion via calcium-sensing receptor (CASR) (237). lipase inhibitor, and arcabose, an ␣-glucosidase inhibitor,
decreased the inhibition of ghrelin secretion by intraduode- changes in food hedonics rather than hunger, a hypothesis
nal lipid infusions and sucrose drinks, respectively (197, supported by neuropharmacological data in animals (211,
248, 250, 749). These studies also revealed that only fatty 370). For example, in rats and mice, injection of ghrelin into
acids with a chain length greater than or equal to C12 the ventral tegmental area, a reward area, activated dopa-
inhibit ghrelin secretion (197, 250). mine neurons, and injection of a ghrelin-receptor antago-
nist into the ventral tegmental area prevented the stimula-
The neuroendocrine reflexes mediating post-meal ghrelin tion of eating by peripheral ghrelin administration (4).
inhibition by intestinal nutrient sensing are poorly under-
stood. The vagus nerve seems unnecessary in rats because Animal studies also link ghrelin signaling to brain networks
vagotomy did not affect post-meal ghrelin inhibition in rats thought to be related primarily to homeostatic eating. For
(830). CCK and PYY(3–36) may be involved because intra- example, in mice, ghrelin administration into the Arc
venous infusions of each reduced plasma ghrelin levels in acutely stimulated eating and altered the activities of Arc
humans (61, 104, 198), whereas GLP-1 infusion did not neuropeptide Y, agouti-related peptide, and pro-opiomela-
(104). We are aware of only one test of the physiological nocortin neurons (145, 164, 810). Ghrelin also appears to
relevance of these potential endocrine controls of ghrelin act in the Arc to reduce serotonin 2C receptor-mediated
secretion: CCKA-receptor blockade abolished long-chain inhibition of eating (661). Finally, initial reports that the
fatty acid-induced ghrelin inhibition in healthy subjects, vagus nerve was required for ghrelin to stimulate eating (36,
suggesting that the mechanism involves CCK (197). Finally, 185, 186) were not replicated when a more selective lesion
although fasting plasma ghrelin levels correlated with basal method was used, which also supports a central action of
leptin levels (240), leptin infusion failed to reinstate normal ghrelin on eating (34).
meal-related ghrelin patterns in healthy-weight men who
had fasted 3 days (139). An unresolved challenge to the hypothesis that ghrelin sig-
nals hunger is that transgenic mice with reduced ghrelin
signaling do not display a tonic increase in eating (524).
C. Eating Some such transgenics do develop obesity, especially when
fed a high-fat diet (524), but this may be secondary to
Changes in plasma ghrelin levels around meals fulfill crite- decreases in fatty acid oxidation and increases in lipid de-
rion 1 of TABLE 2 for an endocrine role in hunger signaling. position in response to changes in autonomic nervous sys-
1) Plasma ghrelin levels increase progressively before meal tem activity (484, 524, 572, 757). As a consequence, ghrelin
onset and fall precipitously afterwards (173, 276, 390, 707, is currently considered to be a stronger candidate for the
778). 2) Hunger ratings were closely related to the drops development of pharmacotherapies for metabolic disease
and subsequent increases of total ghrelin levels between than for overeating.
lunch and a spontaneous dinner in healthy-weight, time-
blinded men (171) as well as between breakfast and a lunch D. Glycemic Control
offered at a set time in overweight and obese men and
women (276). 3) Breakfast-to-lunch intermeal intervals in Ghrelin may affect glycemic control by accelerating gastric
healthy-weight, time-blinded men who were served dinner emptying, inhibiting insulin secretion, or stimulating secre-
upon request were correlated with post-breakfast decreases tion of glucagon or other counterregulatory hormones
in total ghrelin and with the AUC of the breakfast-to-lunch (106, 152, 170, 202, 524, 530, 750, 799). In one study,
ghrelin response (84) [although these correlations were not intravenous infusion of a near-physiological dose of 0.3
detected in non-time-blinded men (124)]. 4) Ghrelin con- pmol·kg⫺1·min⫺1 ghrelin, reduced insulin levels in response
centrations at meal onset correlated with meal size in to intravenous glucose infusion and increased growth hor-
healthy-weight and overweight men and women offered mone and cortisol, but not glucagon, epinephrine, or nor-
lunch at a set time (276). Tests of ghrelin infusions, how- epinephrine, levels (767). Studies in mice indicate that the
ever, have hitherto failed to fulfill criterion 3 of TABLE 2. insulin-inhibitory effect of ghrelin is mediated by a direct
Intravenous infusion of 0.3 pmol·kg⫺1·min⫺1 ghrelin, a action on pancreatic -cells (208, 413). The modulation of
near-physiological dose (TABLE 3), that began 1 h after a ghrelin acylation by dietary levels of C8 and C10 fatty acids
standard meal did not affect subjective hunger, the sponta- may provide a mechanism for brain nutrient sensing and the
neous intermeal interval, or the size of the following spon- neural regulation of glucose metabolism (389), although
taneous meal (444). Pre-meal infusion of 1–5 pmol· given the low levels of these fatty acids in most diets, this
kg⫺1·min⫺1 ghrelin, however, did stimulate eating in two seems unlikely to be a physiological endocrine effect under
tests in which meals were offered at set times (221, 839). most conditions.
Interestingly, supraphysiological ghrelin infusions also in-
creased neural activity in response to pictures of food, as E. Obesity
detected by functional magnetic-resonance imaging (fMRI),
in brain regions associated with food reward (284, 465). GHRL polymorphisms were associated with BMI variation
This suggests that ghrelin may affect eating primarily via in several human populations (430). Although significant,
the effects are quite small [for example, a GHRL polymor- reported that in obese subjects who had elevated fasting
phism at rs35683 accounted for ⬍0.3% of the variance in ghrelin levels and no post-meal ghrelin drops, RYBG ini-
BMI in a sample of 2,000 European-Americans (430)], and tially reduced fasting ghrelin, but that by 1 year post-
the functional pathways that contribute to the effects are RYGB, fasting ghrelin levels were no longer reduced and
unknown. there were typical post-meal drops. Such gradual normal-
ization of ghrelin secretion after RYGB may result either
Fasting plasma ghrelin levels are decreased in obese subjects from weight loss or from dynamic adaptation of the GI tract
and increased by diet-induced weight loss (174, 240, 390, (678). RYGB increased ghrelin levels in some rodent studies
779). Because obesity increases fasting insulin levels, the (31, 780, 854), but decreased them in others (731, 735).
inhibitory effect of insulin on ghrelin secretion (see sect. Stylopoulos et al. (731) suggested that this apparent dis-
IIID) may contribute to obesity’s effect on fasting ghrelin. crepancy may be attributable to an effect of the initial rapid
Shiiya et al. (688), however, did not detect any effect of postsurgical weight loss to increase ghrelin levels combined
T2DM on fasting plasma ghrelin in obese subjects. Post- with a sustained decrease in ghrelin secretory capacity due
prandial drops in plasma ghrelin were reduced in some to the gastric resection. Interestingly, in their rat model,
(239, 240, 497, 574), but not all (103, 174, 403), studies of weight loss 3 months after surgery was correlated with the
obese subjects. pre- to postsurgery decrease in ghrelin levels (731). In an-
other rat study (691), in which there were no consistent
We are aware of one study of the effect of ghrelin on eating changes in pre-meal ghrelin levels tested 12–16 wk after
in obese humans (221), which was inconclusive. Acute in- RYGB, ghrelin levels decreased more after meals in RYGB
travenous infusions of supraphysiological doses of ghrelin than control rats, and the magnitude of the effect was cor-
(1 and 5 pmol·kg⫺1·min⫺1) appeared to increase eating related with weight loss.
more in obese than in healthy-weight subjects, but whether
the differences were statistically significant was not tested.
G. Summary
Stomach
are correlated with hunger sensations and meal size, but if pM at 60 –90 min. Similarly, using the state-of-the-art
ghrelin has a causal endocrine role in hunger is unclear. RAPID method, Eysselein et al. (242) found a 1,600 kcal
Ghrelin may contribute to glycemic control via several mixed-nutrient meal increased plasma CCK from a fasting
mechanisms. Indeed, it has been hypothesized that ghrelin’s level ⬃2.5 to ⬃7 pM at 60 min. A number of studies involv-
major function is to prepare the organism for the nutrient ing isoenergetic loads of highly digestible nutrients that
repletion and storage (389, 524). Studies to date of ghrelin were infused intraduodenally to control gastric-emptying
physiology in obese individuals and after RYGB have not effects indicate that, with respect to both peak values and
produced consistent results. Ghrelin antagonists and in- AUC, 1) oral lipids stimulate CCK secretion most per kcal,
verse agonists suitable for human use (78, 125) may soon proteins are intermediate, and carbohydrates stimulate
resolve many of these outstanding questions. CCK secretion least; and 2) plasma levels increase in 10 –15
min (327, 337, 446, 584, 641, 682).
IV. CHOLECYSTOKININ
Hydrolysis of proteins and triglycerides is required for nor-
mal CCK secretion (55, 159, 247, 325, 479, 718). Addi-
A. Introduction tionally, fatty acids with chain length greater than or equal
to C12 stimulate CCK secretion much more than fatty acids
CCK cells are open-type cells, i.e., their apical surfaces are less than C12 (249, 479, 486, 487), and less saturated long-
exposed to the intestinal lumen (FIGURE 6B). Initial electron chain fatty acids stimulated CCK secretion more than
microscopy and immunocytochemistry studies suggested highly saturated fatty acids (67). Carbohydrate digestion
that they were a unique species of enteroendocrine cells, may not be required, as the ␣-glucosidase inhibitor acar-
called I-cells (590). Contemporary methods, however, indi- bose had little or no effect on the CCK response to mixed-
cate that, at least in rodents, many enteroendocrine CCK nutrient meals (236, 751, 784).
cells also express and secrete ghrelin, GLP-1, PYY, GIP,
neurotensin, or secretin (231, 303, 597, 738, 742). In hu- Consistent with the higher density of enteroendocrine CCK
mans, swine, and rats, enteroendocrine CCK cells are cells in the proximal small intestine, intraduodenal glucose
densely expressed in the duodenum and proximal jejunum, infusions that were prevented from transiting more than 60
less dense in the distal jejunum, and sparse in the ileum (45, cm distal to the pylorus by an inflated balloon stimulated
478, 503). CCK secretion as much as infusions done without balloon
inflation (445). This is likely also to be the case for fat and
CCK circulates predominantly in a 58-amino acid form protein. Intraileal lipid infusion also increased CCK secre-
(CCK-58) (243, 431, 612, 722). Importantly, many CCK tion (466), but whether this was due to a direct action on
assays that involve plasma formation recover ⬍20% of en- ileal CCK cells or to an indirect, presumably endocrine,
dogenous CCK, so they provide accurate relative, but not distal-to-proximal reflex is unknown.
absolute, levels (243, 431, 722). Additionally, most tests of
exogenous CCK use CCK-8, which is rare or absent in the Intraluminal nutrients directly and indirectly stimulate
plasma. This may be important because the liver clears CCK secretion. Direct nutrient effects are mediated by a
CCK-8 faster than larger forms (287, 404) and CCK-8 had variety of nutrient receptors expressed on the apical surface
slightly different effects than CCK-33 or CCK-58 in animal of CCK cells (FIGURE 6B AND TABLE 4, which includes the
models (607, 608), including in tests of eating in rats (232, full and the former names of the nutrient receptors dis-
279, 281). cussed below). In humans, free fatty acids act on FFAR1
(443), FFAR4 (752), and the fatty-acid transporter CD36
There are two CCK receptors, CCKAR (or CCK1R) and
(733); oligopeptides and amino acids act on CASR (161,
CCKBR (CCK2R) (216, 514, 515, 612). CCKAR is more
328, 811), LPAR5 (149), TAS1R1/TAS1R3 (160, 182,
abundant peripherally than centrally and requires the seven-
543) and, perhaps, SLC15A1 (183). The presence of tran-
amino carboxy-terminal segment and sulfation of the ty-
scripts for TAS1R2/TAS1R3 and GNAT3 on CCK-secret-
rosine residue at position 7 for full activation. CCKBR, or
ing mouse enteroendocrine STC-1 cells suggests that sweet-
the gastrin receptor, is sensitive to unsulfated CCK hexa-
receptor signaling may contribute to glucose-induced CCK
peptides and is abundant both peripherally and centrally,
release in mice (228, 849). This may not be the case in
where CCK-8 is a neurotransmitter.
humans, however, because intragastric and intraduodenal
infusions of the sweet-receptor inhibitor lactisole that re-
B. Secretion duced glucose-induced GLP-1 and PYY secretion did not
affect CCK secretion (275). Intraluminal nutrients also
Mixed-nutrient meals increase CCK secretion. Using a well stimulate CCK secretion indirectly via the CCK-releasing
validated radioimmunoassay, Rehfeld et al. (609) found factors “pancreatic monitor peptide” and “intestinal lumi-
that a 1,470 kcal mixed-nutrient meal increased plasma nal CCK-releasing factor” (456, 504, 812). This occurs in
CCK from a fasting level of ⬃1 to ⬃3 pM at 30 min and ⬃5 part due to binding of proteases to proteins and lipids,
which reduces protease-induced degradation of CCK-re- obesity (192, 473, 501), suggesting that endogenous CCK is
leasing factors (168, 432). also important for the tonic control of eating. 2) fMRI
following intragastric lauric acid loads with or without
loxiglumide indicated that CCK signaling is crucial for nor-
C. Eating
mal brain responses to this fatty acid (419) (because lauric
CCK is the best-established GI endocrine satiation signal in acid is uncommon in Western diets, the generality of this
humans. First, in three studies (54, 299, 438), intravenous finding is uncertain). 3) CCK doses substantially above
infusions of physiological doses of CCK reduced meal size physiological (i.e., ⬃1.8 to ⬃3.5 pmol·kg⫺1·min⫺1, TABLE
3) inhibited eating without eliciting adverse effects (100,
without adverse physical or subjective effects in men and
women, which fulfills criteria 3 and 5 of TABLE 2. The study 269, 290, 391). Interestingly, CCK infusions reduced meal
by Lieverse et al. (438) is especially interesting because the size ⬃30 –50% in these studies without affecting fullness or
test food, bananas, did not elicit CCK secretion under their other meal-related sensations compared with the control
conditions (440), so that the infused CCK did not synergize condition, suggesting that CCK had an effect on conscious-
with endogenous CCK, as probably happens in most satia- ness indistinguishable from the presumably more complex
tion tests. Second, intravenous infusions of the CCKAR afferent activation produced by the larger quantity of food
antagonist loxiglumide increased premeal hunger feelings, eaten in the control condition. 4) In most of these studies,
reduced fullness feelings during the meal, increased meal CCK infusions began after a small preload to capitalize on
size, and blocked the satiating effects of intraduodenal lipid the synergy between gastric mechanoreception and CCK
infusion (70, 439, 480), which fulfills criteria 4 and 6 of (528), described in section IIC.
TABLE 2. These studies, summarized in FIGURE 8, have
made CCK paradigmatic for the study of the endocrine Attempts to relate endogenous CCK levels with subjective
control of eating. measures of appetite have been less informative than studies
of manipulation of CCK. 1) In the sole study of intrameal
In addition, 1) human CCKAR polymorphisms are associ- effects, plasma CCK increased more during meals in women
ated with increased meal size, increased food intake, and than in men, but hunger and fullness ratings did not differ;
A g B kcal C kcal
500 2500 2500
*
400 2000 2000 *
+
*
Meal Size
Meal Size
Meal Size
0 0 0
IV SAL CCK IV SAL SAL LOX IV SAL LOX
ID SAL FAT FAT
FIGURE 8. Evidence that endogenous CCK signals satiation in healthy humans. A: intravenous infusion of a
physiological dose of CCK inhibited eating. Ten healthy-weight women [body mass index (BMI) 22 ⫾ 3 kg/m2]
and 8 obese women (BMI 39 ⫾ 2 kg/m2) received 60 min intravenous (IV) infusions of 0.24 pmol·kg ideal body
weight⫺1·min⫺1 CCK-33 or saline (SAL) beginning at 0800 after an overnight fast. At 0900, a 132 kcal
preload of bananas was served, and at 0915, a banana-shake meal was served in excess; bananas were used
because they did not elicit CCK secretion. CCK significantly reduced meal size (*) without physical or subjective
side effects. [From Lieverse et al. (438), with permission from BMJ Publishing Group Ltd.] B: the CCKA
receptor antagonist loxiglumide (LOX) antagonized the satiating action of endogenous CCK stimulated by
intraduodenal (ID) infusion of a fat emulsion. Healthy-weight adult males began a midday lunch buffet 4 h after
a standard breakfast, 90 min after onset of an IV infusion of 10 mol·kg⫺1·h⫺1 LOX or SAL, 60 min after an
ID infusion of 0.4 ml/min corn oil (FAT) or SAL, and 20 min after an oral preload of 400 ml of a low-fat banana
milkshake. Infusions were continued throughout the meal. ID fat infusion significantly reduced the size of the
lunch meal (⫹), and that this was reversed by LOX (*); no physical or subjective side effects occurred in any
condition. [From Matzinger et al. (480).] C: antagonism of CCK signaling with the CCKA receptor antagonist
LOX stimulated eating. Healthy-weight adult males began a midday lunch buffet 4 h after a standard breakfast
and 60 min after beginning an IV infusion of 22 mol·kg⫺1·h⫺1 LOX or SAL. Infusions were continued
throughout the meal. LOX significantly increased meal size (*) without physical or subjective side effects. [From
Beglinger et al. (70).]
women gave higher ratings of “sickness” early in the meal, the satiating effect of exogenous CCK (516). As little or no
but not later when CCK levels increased more, nor did they food reaching the pylorus is digested sufficiently to stimu-
spontaneously report illness or display signs of illness (549). late CCK secretion and the CCKAR are localized in the
The small sample size (four of each sex) further limits this muscle layer rather than the mucosal layer (565), the pyloric
study. 2) Postprandial CCK levels and hunger and fullness contribution to CCK satiation is likely to be endocrine. 2)
ratings were significantly correlated in a group of nine men, Vagotomy studies suggest that endocrine CCK may also act
but the relationships were not detected in all individuals (3 in the brain to inhibit eating (615, 850). For example, in-
of 9 for hunger and 4 of 9 for fullness) (260). 3) Meals travenous infusions of devazepide, which enters the brain,
containing different fats differentially increased postpran- stimulated eating after vagotomy, whereas infusions of a
dial plasma CCK levels in eight women, and these were larger-molecule CCKAR antagonist that penetrates periph-
mirrored by subjective hunger and fullness ratings; but nei- eral capillaries, but not the blood-brain barrier, did not
ther CCK responses nor appetite ratings differed in seven (615). The site of the brain CCKAR mediating these effects
men (119). 4) Meals containing different fat-to-carbohy- is not known. The NTS (83, 330), to which vagal afferents
drate ratios differentially increased postprandial plasma project, and the dorsomedial nucleus of the hypothalamus
CCK levels in 16 overweight and obese men and women, (79) are candidates.
but no associations with subjective appetite were detected;
there was also no difference in the size of meals offered 3 h Whether CCK’s physiological satiating effect in humans
later, but by this time CCK levels had returned to basal involves local or endocrine action is unclear. That infusions
(277). Because CCK appears to signal satiation, but not mimicking systemic levels reached during meals are suffi-
postprandial satiety, it is unfortunate that there are not cient to reduce eating even when endogenous CCK secre-
more studies of the relationships among differential in- tion is minimized (438, 440) suggests, but does not prove,
trameal plasma CCK levels, appetite, and meal size. that local signaling is not responsible. This is because GI
hormones diffuse down a steep concentration gradient from
Reproductive physiology may affect CCK satiation. the lamina propria into the mesenteric veins and are then
Women spontaneously eat progressively less during the fol- successively diluted in the hepatic-portal circulation and
systemic circulation (FIGURE 3). Thus, although the exact
licular phase of the menstrual cycle, reaching a nadir in
difference between lamina propria and systemic CCK con-
daily food intake during the periovulatory phase that is
centrations is unknown, it seems likely that physiological
⬃275 kcal/day less than the luteal-phase maximum (38).
intravenous CCK infusions are not sufficient to reproduce
Rats and mice also display a decrease in food intake during
the CCK concentrations in the lamina propria that occur
the periovulatory phase, due in part to an increase in the
during meals.
satiating potency of CCK related to estrogen signaling in the
nucleus of the solitary tract (NTS) (38).
D. Glycemic Control
Studies in rodents suggest that CCK inhibits eating via both
local and endocrine modes of action. In support of local No direct role has been established for CCK in glycemic
action, intravenous infusion of the small-molecule CCKAR control in humans. Infusion of a physiological CCK-8 dose
antagonist devazepide, which presumably diffused from the (0.4 pmol·kg⫺1·min⫺1) reduced plasma glucose after an
capillaries into the small intestinal-lamina propria, in- oral glucose load, but not after an intraduodenal glucose
creased food intake, but infusions of a CCK antibody, infusion that mimicked the gastric-emptying rate of the oral
which would not escape the vasculature so as to selectively glucose (437), suggesting that CCK reduces blood glucose
block endocrine effects, did not (616). Infusion studies in- indirectly via inhibition of gastric emptying. In two studies,
dicate that the most likely physiological site of CCKAR however, the CCKAR antagonist loxiglumide failed to af-
mediating satiation is the proximal small intestine (165, fect plasma glucose despite accelerating gastric emptying
814). In addition, the satiating action of intraperitoneal (263, 323). Physiological levels of CCK do not appear to
injections of CCK is mediated by vagal afferent fibers (165, lower blood glucose by increasing insulin secretion because
426, 704, 705), and most small intestinal vagal afferents infusion of 0.2– 0.4 pmol·kg⫺1·min⫺1 CCK-8 did not in-
terminate within the crypt and villous lamina propria, but crease the insulin response to co-infusion of glucose (637).
not in close apposition to enteroendocrine CCK cells, indic- Physiological infusions of CCK did, however, increase the
ative of a paracrine action (77). Nevertheless, some vagal insulin response produced by amino-acid infusions in two
afferents terminate with 5 m of CCK cells (77), and CCK (324, 637) of three (253) studies. But several attempts to
neuropods appear to signal via enteric glial cells (90) (de- demonstrate a direct insulinotropic effect of CCK using
scribed in section IB3 and FIGURE 3) so that neurocrine or various CCKAR antagonists and various nutrient stimuli
neuropod satiation signaling is also possible. failed (65, 263, 323, 324, 400, 434, 547, 667, 673).
Other data in rats and mice support an endocrine mode of CCK secretion was reduced in patients with longstanding
action. 1) CCKAR in the pyloric muscle layers contribute to T2DM (112, 636), perhaps due to the autonomic neuropa-
thy and reduced rates of gastric emptying typical of these the satiating effect of exogenous CCK, and the de-satiating
patients (344). CCK-8 infusion further slowed gastric emp- effect of devazepide were reduced in rats fed a high-fat diet
tying and improved postprandial insulinemia and glycemia (225, 739).
in patients with T2DM (18, 580, 636). Thus CCK agonists
may be useful in diabetes therapy. Reports that rats that received intraperitoneal infusions of
CCK during every spontaneous meal for 6 days (821) and
Studies in rats indicate that CCK affects glucose metabolism mice with transgenic null mutations of Cckar (402) in-
by reducing hepatic glucose production via a vagal-vagal creased meal frequency and failed to gain weight contrib-
reflex (98, 99, 147, 605). CCK was infused intraduodenally uted to the views that CCK (and by extension other GI
in amounts that failed to increase CCK concentration in the hormones) does not contribute significantly to tonic energy
hepatic-portal vein to mimic the local action of CCK in the homeostasis and is a poor candidate for obesity pharmaco-
lamina propria, thus providing unique evidence for a para- therapy. [The OLETF rat, which also bears a Cckar null
crine action. This method seems feasible for human studies mutation, is obese, but this is apparently due to loss of
and may lead to better understanding of the relative roles of
hypothalamic Cckar rather than vagal Cckar (79).] Recent
local and endocrine signaling in GI hormone function.
preclinical data are more promising. First, stabilized forms
of CCK that resist enzymatic degradation reduced food in-
E. Obesity take and body weight in various mouse obesity models
(357, 359, 582). Second, native and stabilized CCK and
Whether obesity affects CCK secretion is controversial. CCKAR agonists increased the anorectic and weight-low-
Fasting CCK levels were reduced in obese subjects in one ering actions of a GLP-1 receptor agonist, amylin, leptin, or
study (57), but not two others (103, 725). CCK responses to amylin plus leptin in various rat and mouse obesity models
intraduodenal oleic acid infusions tended to be delayed and (358, 776, 777), as did a CCK/GLP-1 agonist hybrid pep-
reduced in overweight or obese compared with healthy- tide (360). These promising results suggest that increased
weight subjects in one study (725), but CCK responses to CCK signaling may contribute effectively to obesity phar-
high-fat, high-carbohydrate, and high-protein meals were macotherapy.
comparable in obese and healthy-weight subjects in another
(103), and CCK responses to ingestion of high-fat meals
were larger in obese than healthy-weight subjects in a third F. RYGB
study (261). Whether these contrasting results were due to
differences in the specific nutrient stimuli used, in gastric CCK has not been a focus of RYGB research because RYGB
emptying, which was not assessed, or other factors is not prevents ingesta from contacting the majority of CCK-se-
known. creting cells. Nevertheless, intraileal lipid infusions in-
creased CCK secretion in healthy-weight subjects (466),
Some defects in CCK signaling can lead to obesity. As men- and postprandial CCK levels were normal (383, 622, 634)
tioned above, human CCKAR polymorphisms are associ- or increased (364, 574) following RYGB. For example, Pe-
ated with increased meal size, increased food intake, and terli et al. (574) found that CCK levels 30 min after a mixed-
obesity (192, 473, 501), suggesting that CCK-signaling de- nutrient meal were increased approximately twofold 1 wk,
fects can contribute to obesity etiology. In addition, allelic 3 mo, and 1 yr after RYGB. That the increase occurred just
variations in CCK were significantly more prevalent in
1 wk post-RYGB suggests that it does not require prolifer-
obese persons who habitually ate very large meals than
ation of CCK cells. The larger increases at later points may
those who did not (the “extreme discordant phenotype”
be related to the proliferation of CCK cells, which was
approach) (192).
reported in the Roux and common limbs of RYGB rats
We know of only one study comparing the satiating action (525). The effect of RYGB on CCK secretion is an interest-
of CCK infusions in healthy-weight and obese humans ing and under-researched phenomenon that may lead to
(438). No difference was obtained (infusion of 0.24 new opportunities for obesity therapy.
pmol·kg⫺1·min⫺1 CCK-33 reduced meal size ⬃18% in 10
healthy-weight women and ⬃20% in 8 obese women). RYGB reduced food intake and body weight in obese
OLETF rats (305), indicating that Cckar signaling is not
Obesity produced by high-fat feeding may interfere with necessary for some response to RYGB in this model. As no
CCK satiation. 1) CCK injections and balloon distension in ad libitum-fed, genetically normal RYGB rats were in-
isolated jejunal segments elicited smaller vagal-afferent cluded in the experiment, however, it is unclear whether the
electrophysiological responses in mice made obese by feed- OLETF rats were normally responsive to RYGB. We know
ing a high-fat diet than in chow-fed mice (181). 2) The CCK of only one test of acute CCKAR antagonism in RYGB rats
responsivity of vagal afferents was reduced in high-fat fed, (37), which failed to reveal any effect of RYGB on endoge-
leptin-resistant rats (193). 3) Fat-induced CCK secretion, nous CCK satiation.
V. GLUCAGON-LIKE PEPTIDE-1
A. Introduction
3 Gastric emptying
GLP-1 is secreted by open-type enteroendocrine cells (FIG-
URE 6B), originally identified as L cells (114, 589), located
in both the small and the large intestine (336, 597). Most
GLP-1 cells in the distal jejunum and ileum coexpress and
secrete PYY; in addition, some GLP-1 cells coexpress CCK,
GIP neurotensin, or secretin (231, 303, 568, 597, 738,
742). GLP-1 is also produced by a small group of neurons
located in the NTS (72, 128, 222, 336). Two equipotent
molecular forms circulate, GLP-1(7–37) and GLP-1(7- FIGURE 9. Some features of CCK physiology. CCK (red dots) se-
36NH2); the latter predominates in humans. Dipeptidyl cretion is stimulated by the digestive products of all three macronu-
peptidase-4 (DPP-4), a proline/alanine-specific peptidase trients acting on nutrient receptors on the apical aspects of en-
found on the luminal surface of capillary endothelial cells, teroendocrine CCK cells (blue) dispersed in the epithelial layer (tan)
of the small intestinal mucosa. CCK acts in an endocrine mode by
in the liver, and in the blood, rapidly degrades active GLP-1 diffusing through the lamina propria (yellow) and into intestinal cap-
to inactive forms, GLP-1(9 –37) and GLP-1(9-36NH2). In illaries (salmon) to reach distant target organs (red arrows), or acts
swine, only ⬃25% of active GLP-1 secreted from the intes- locally. 1) CCK’s physiological effects include stimulating satiation.
tine reaches the portal circulation, and only ⬃10 –15% This may occur via endocrine actions in the pyloric area of the
reaches the systemic circulation (336). stomach that produce signals relayed to the brain via vagal afferents
(green arrow) or via local actions on vagal afferents in the lamina
propria. An endocrine action in the brain may also contribute. 2)
GLP-1 receptors (GLP-1R) are expressed in the GI tract, CCK lowers meal-related glycemia via an endocrine effect on gastric
pancreas, cardiac atrium, abdominal vagal afferents, and emptying and perhaps via a vagal-vagal reflex. 3) Similarly, CCK
many brain areas (28, 115, 128, 336, 450, 623, 783). In slows gastric emptying via a direct endocrine effect and perhaps via
some animals, GLP-1 degradation products appear to sig- a vagal-vagal reflex. Solid lines indicate well-established effects, and
dashed lines indicate less well established effects.
nal via non-GLP-1 receptors (766). Whether these peptides
have physiological functions in humans is not known.
the morning level between meals (130, 235, 322, 557, 681,
804, 805). For example, when healthy-weight men and
B. Secretion women ate 524 kcal breakfasts and, 4 h later, mixed-nutri-
ent lunches containing 511, 743, or 1034 kcal, active
Available GLP-1 assay methods usually yield similar rela- GLP-1 increased from ⬃5 pM to ⬃8, ⬃12, and ⬃16 pM,
tive changes, but often different absolute concentrations respectively, after 30 min and then decreased to ⬃7 pM
(49, 410). Due to the rapid degradation of active GLP-1, after 180 min (27).
GLP-1 secretion is best estimated by the sum of active and
inactive GLP-1 in plasma (336, 508). Plasma GLP-1 con- Oral loads of glucose and several other carbohydrates usu-
centrations are at their lowest levels after overnight fasts, ally result in monophasic increases in plasma GLP-1, with
increase rapidly during meals, and usually do not return to onsets after 5–15 min, peak values after 15–30 min, and
initial values regained after 3– 4 h (130, 235, 322, 399, 663, gastric empting contributes to the control of GLP-1 secre-
714, 759, 793, 805). Oral fructose, however, was only tion.
about half as potent a GLP-1 secretogogue as oral glucose,
both on a molar basis (409) and when matched for per- The distal small intestine usually plays the leading role in
ceived sweetness (716). Oral protein and lipid typically pro- sustained GLP-1 secretion. This was first indicated by com-
duce slower-onset, more sustained increases than does glu- parisons of GLP-1 secretion and glucose absorption. The
cose (95, 122, 128, 131, 195, 235, 336, 442, 600, 762). Due threshold intraduodenal glucose infusion rate for sustained
to the complex patterns of GLP-1 secretion described be- increases in plasma GLP-1 was between 1 and 2 kcal/min in
low, however, it is unclear whether any macronutrient two studies (663, 774) and between 2 and 4 kcal/min in
should be considered to be the most potent GLP-1 secreta- another study (583). Because the absorptive capacity of the
gogue. Both fasting and glucose-stimulated GLP-1 secretion duodenum and first 25–30 cm of jejunum is ⬃0.9 –1.4 kcal/
are pulsatile, with a period of ⬃8 min (50); the controls and min (334, 505, 575), glucose probably reached the more
consequences of this are not known. distal jejunum in these studies only when at least ⬃1.5
kcal/min glucose was infused, which suggests that stimula-
GLP-1 levels after mixed-nutrient meals sometimes display tion of more distal GLP-1 cells is required to elicit sustained
biphasic patterns (130, 235, 604), with secondary peaks GLP-1 secretion. Two experiments confirm this suggestion.
after 60 –120 min. Differences in rates of gastric emptying The first (159) involved intraduodenal infusion of 3.5 kcal/
and other differences in digestibility of the meal compo- min glucose 2 cm distal to the pylorus, aspiration of the
nents are likely to contribute to this, but are not their sole intestinal contents 60 cm distal to the pylorus, inflation of
cause because biphasic patterns also occurred after oral an occluding balloon just distal to the aspiration site, and
loads of individual nutrients (130, 322). intrajejunal infusion of glucose or saline distal to the occlu-
sion, 75 cm distal to the pylorus. Plasma GLP-1 levels in-
There may be sex-specific effects on GLP-1 secretion. Oral creased when glucose was infused intrajejunally in amounts
glucose and mixed-nutrient meals increased GLP-1 levels matching the aspirated glucose, but not when saline was
more in women than men in two studies (763, 805), but not infused intrajejunally. In the second (844), 2 kcal/min glu-
in two others (132, 385). In addition, oral glucose increased cose was infused either via intraduodenal catheters that
GLP-1 levels less during the follicular phase than during the ended 12 cm distal to the pylorus or via intrajejunal cathe-
luteal phase, apparently due to slower gastric emptying ters that ended 50 cm distal to the pylorus; in this condition
(101). a balloon was inflated 30 cm distal to the pylorus to exclude
reflux. Plasma GLP-1 levels increased markedly more when
Several mechanisms may contribute to the rapid initial glucose was infused intrajejunally than intraduodenally.
GLP-1 response: 1) GLP-1-cells are expressed in the duode- These two experiments demonstrate that small intestinal
num and proximal jejunum (148, 365, 738, 759), so direct glucose stimulation ⱖ50 –75 cm distal to the pylorus is
stimulation of GLP-1 secretion occurs as soon as ingesta necessary for GLP-1 secretion and that small intestinal glu-
pass the pylorus. 2) The initial rate of gastric emptying of cose stimulation ⬍50 – 60 cm distal to the pylorus is not
glucose solutions, especially in fasted subjects, may produce sufficient for it.
glucose concentrations that exceed the absorption capacity
of the proximal small intestine, so that glucose may reach Further observations also attest to the importance of distal
more distal GLP-1 cells within 5–10 min after meals (50, small intestinal GLP-1 cells in GLP-1 secretion. 1) Reducing
663). 3) The rate of increase in plasma GLP-1 may exceed intestinal nutrient transit with hyoscine decreased GLP-1
the rate of increase of plasma glucose, suggesting that neu- secretion in response to glucose (137). 2) GLP-1 secretion
roendocrine reflexes may stimulate GLP-1 secretion in ad- was increased when carbohydrates were administered with
dition to direct stimulation of GLP-1 cells by glucose (508). the acarbose, which slows digestion of starch and disaccha-
rides and increases the amounts of carbohydrates reaching
Intraduodenal-infusion studies, similar to oral-loading the more distal small intestine (274, 601, 680, 784, 785). 3)
studies, indicate that carbohydrate increases plasma GLP-1 GLP-1 secretion was increased more by lower glycemic load
levels faster than either protein or lipid (71, 247, 249, 412, foods, i.e., less digestible, than by higher glycemic load
446, 550, 583) and that biphasic responses sometimes oc- foods (635), again presumably by increasing the amounts of
cur (446, 583). In addition, 1) for each macronutrient, in- carbohydrates reaching the more distal small intestine. 4)
creasing caloric load increased GLP-1 levels (446, 583, Delivery of small amounts of lauric acid to the ileum and
640). 2) GLP-1 responses were greater during initially faster colon by enteric-coated pellets increased meal-induced
and subsequently slower nutrient infusions compared with GLP-1 secretion (459). 5) GLP-1 responses to intraduode-
identical loads infused at constant rates, suggesting that nal infusions of amino acids and fatty acids are slow in
increases in the initial intraluminal nutrient load dispropor- onset (71, 718), which, because no digestion is required in
tionally increase GLP-1 secretion (136, 550). Because gas- these situations, indicates that GLP-1 cells in the proximal
tric emptying rates are highest initially, this suggests that small intestine are not sufficient for the responses. 6) That
GLP-1 cells are more dense in the distal jejunum and ileum receptor FXR appears to inhibit GLP-1 production because
than in the more proximal small intestine in humans (233) treatment with a bile-acid sequestrant improved glucose
also supports the importance of the distal small intestine in tolerance and increased ileal GLP-1 expression in wild-type
GLP-1 secretion. mice, but not Fxr knockout mice (773). The importance of
bile acids for human GLP-1 secretion is not clear. 1) Bile
Many products of digestive hydrolysis directly stimulate acid and GLP-1 responses were correlated in one study
GLP-1 secretion via membrane receptors on the apical sur- (627). 2) Intrajejunal infusion of taurocholic acid did not
faces of enteroendocrine GLP-1 cells (TABLE 4, which in- affect GLP-1 secretion by itself, but increased glucose-stim-
cludes the full and the former names of the nutrient recep- ulated GLP-1 secretion beginning after ⬃90 min (841). 3)
tors discussed below). Intragastric infusion of lactisole, an Intraduodenal infusion of chenodeoxycholic acid had only
inhibitor of the TAS1R2/TAS1R3 sweet receptor, reduced a small effect on plasma GLP-1 levels (496).
the GLP-1 response to intragastric glucose in humans (275,
717), suggesting that GLP-1 cells express these receptors There appear to be several reflexive controls of GLP-1 se-
(228). TAS1R2/TAS1R3 receptors do not appear to be suf- cretion. 1) The nicotinic cholinergic antagonist trimeth-
ficient for GLP-1 secretion, however, because artificial aphan did not reduce the early increase in GLP-1 after a
sweeteners that stimulate them had no effect (716, 845, mixed-nutrient meal, but did reduce the increases in insulin
849). Glucose absorption via the glucose transporters and pancreatic polypeptide, suggesting that cephalic-phase
SLC5A1 and SLC2A2 may be required because selective reflexes did not contribute to the GLP-1 response (17). A
inhibitors of both transporters reduced GLP-1 secretion in preprandial cephalic-phase GLP-1 reflex, however, was
animals (461, 519). SLC2A1 and SLC2A5 have also been demonstrated in rats (180, 786). 2) The muscarinic cholin-
found on GLP-1 cells (618); the latter suggests a mechanism ergic antagonist atropine reduced the GLP-1 response to an
for the stimulation of GLP-1 secretion by fructose men- oral glucose load (50). 3) Mouse GLP-1 cells express the
tioned above. melanocortin 4 receptor (Mcr4), whose activation in-
creased GLP-1 secretion (561).
There is indirect evidence that protein hydrolysis is required
for GLP-1 secretion (142, 302, 322, 461, 718, 760). Oligo- The afferent limbs of GLP-1 reflexes may involve gastric-
peptides and amino acids may be sensed by CASR (210, phase signals because intragastric infusion of glucose or a
461). Phenylalanine, tryptophan, asparagine, arginine, and mixed-nutrient solution led to greater plasma GLP-1 levels
glutamine each stimulated GLP-1 in isolated rat small intes- than matched intraduodenal infusions (719), although it is
tine, and this was abolished in the absence of extracellular also possible that the initial rate of gastric emptying may
Ca2⫹ or the presence of a CASR inhibitor (461). have exceeded the rate of intraduodenal infusions, leading
to greater direct stimulation of GLP-1 release. An intestinal-
As for ghrelin and CCK, the effect of lipids on GLP-1 secre- phase reflex appears to contribute to GLP-1 secretion in
tion depends on digestive hydrolysis and the presence of response to intraduodenal fat infusions because GLP-1 did
fatty acids with chain length greater than or equal to C12 not increase after CCKAR antagonism (71). Finally, re-
(71, 249). The free fatty-acid receptors FFAR2, FFAR3, and search in rats indicates that vagal signaling contributes im-
FFAR4 are densely expressed on distal small intestinal portantly to reflexive GLP-1 release (29, 110, 628).
GLP-1 cells (329, 380, 715, 754), which may contribute to
the slower-onset, more sustained GLP-1 responses after
lipid ingestion. Carbohydrates reaching the large intestine C. Eating
are fermented to short-chain fatty acids, which are sensed
by FFAR2 and FFAR3 (206, 617) and contribute to the GLP-1 fulfills criterion 3 of TABLE 2 for an endocrine sati-
later phase of GLP-1 secretion (376, 765). For example, ation signal in humans because intravenous infusion of
oral loads of xylose, a poorly absorbed sugar, led to greater physiological doses of GLP-1 (0.3– 0.9 pmol·kg⫺1·min⫺1)
increases in GLP-1 than did glucose from 60 to 180 min reduced meal size in the absence of adverse effects in
after loading (793). GLP-1 cells also express GPR119, healthy-weight men (199, 255, 299, 301). Test context may
which mediates responses to oleoethylamine (151, 420, affect GLP-1 satiation, however, because a physiological
618), a long-chain fatty acid derivative formed during ab- dose failed to inhibit eating in another test (100), and sup-
sorption. raphysiological doses (1.2–1.5 pmol·kg⫺1·min⫺1 GLP-1)
reduced meal size in one (301), but not in another study
Nutrients may also stimulate GLP-1 secretion indirectly by done under different conditions (454). The site(s) of the
increasing bile secretion. Bile acids appear to control GLP-1 GLP-1R mediating this satiating effect is unknown. As ex-
secretion in two ways. First, circulating bile acids diffuse plained in section IVC, we conclude that positive results
into the lamina propria and reach GPBAR1 on the basolat- with physiological endocrine doses suggest that GLP-1 acts
eral aspects of GLP-1 cells, which stimulates GLP-1 secre- via an endocrine mode of action, rather than signaling lo-
tion in mice (105, 564, 761). Second, the nuclear bile-acid cally, to inhibit eating in these tests.
With respect to criterion 6 of TABLE 2, GLP-1R antagonism Animal studies have also identified a number of brain sites
with exendin(9 –39) failed to increase test meal size in where GLP-1 may act to inhibit eating, including the area
healthy humans tested under five distinct experimental con- postrema, NTS, lateral parabrachial nucleus, ventral teg-
ditions (492, 721) and in two groups of RYGB patients mental area, paraventricular nucleus of the hypothalamus,
tested 3–12 mo after surgery, when their mean BMI was and nucleus accumbens (23, 219, 499, 500, 599, 619, 620,
⬃34 kg/m2 (737). In contrast, exendin(9 –39) did increase 652). Because the data reviewed above suggest that GLP-1
test meal size in one of the RYGB-patient groups in a pre- does not increase in the systemic circulation after meals in
operative test (737). Several factors may have contributed rats and that intestinal GLP-1 does not control eating in rats
to these disparate results. 1) The positive result was ob- via an endocrine mechanism, however, these sites are prob-
tained with a primed infusion and a higher maintenance ably physiologically stimulated by neuronal GLP-1 in rats,
dose of exendin(9 –39). 2) Meal-related GLP-1 secretion which originates in neurons in the caudal brain stem that
was markedly increased in the patients tested after RYGB, project to all the areas listed above and more (450, 623).
so even the higher exendin(9 –39) dose may not have been Because postprandial GLP-1 levels are relatively high in the
sufficient. 3) Patients were heavier before RYGB (⬃40 kg/ systemic circulation in humans, it is possible that endocrine
m2) than after (⬃34 kg/m2), suffered from T2DM, and were GLP-1 does affect these brain areas in humans. GLP-1 ap-
receiving insulin treatment (after RYGB only 2 of 12 pa- pears to enter the brain by simple diffusion (381). Finally,
tients had T2DM, and treatment was not specified). 4) Ex- rat studies indicate that GLP-1 signaling in the NTS affects
endin(9 –39) led to unusually high levels of PYY(3–36) in satiation in part by modulating the processing of signals
healthy subjects (721) and in patients tested after RYGB, related to gastric fill (312, 806), although whether endo-
but failed to increase PYY(3–36) in patients tested before crine or neurocrine GLP-1 is involved is unclear.
RYGB (737).
GLP-1 may contribute to postprandial satiety as well as to
Intravenous GLP-1 infusions failed to inhibit eating in men satiation. 1) As described above, GLP-1 levels are often
with truncal vagotomy and pyloroplasty (588), suggesting increased for several hours postprandially. 2) In a test of
that GLP-1 acts in the abdomen to inhibit eating. Many healthy-weight subjects who consumed fixed-size, high-fat,
data in rats also support a vagal mechanism (1, 313, 407, low-carbohydrate test meals (276), plasma GLP-1 levels
639), although capsaicin lesions of unmyelinated abdomi- 60 –180 min later were correlated with both hunger ratings
nal afferents failed to block GLP-1’s eating-inhibitory effect and the size of meals offered at 180 min (neither correlation
in one study (613). was present 0 – 60 min after the fixed-size meals). 3) In rats,
chronic intrajejunal infusions of linoleic acid increased
Additional rat and mouse data also indicate that intestinal GLP-1 levels and selectively reduced meal frequency, and
GLP-1 acts locally to inhibit eating in these species. 1) intraperitoneal exendin(9 –39) infusion reversed the reduc-
Meals failed to increase systemic active GLP-1 levels in rats tion in meal frequency (179).
(598, 691) [although meals did lead to GLP-1 increases in
mice (15, 297)]. 2) Infusion of 0.5 nmol/kg GLP-1 via the GLP-1 may affect eating in other ways. 1) GLP-1 may con-
cranial-mesenteric artery, which supplies much of the small tribute to flavor hedonics (24, 212, 499, 699, 827). Many of
and large intestines, reduced meal size more than identical the effects on rats’ eating produced by central GLP-1 ma-
infusions via the hepatic-portal vein or femoral artery in nipulations described above related to “hedonic eating,”
rats (832). 3) Hepatic-portal vein infusion of 1 nmol/kg and the effects of GLP-1 on human fMRI responses to pic-
GLP-1, which was near the threshold for an eating-inhibi- tures of foods also occurred in brain areas related to the
tory effect under the conditions tested in rats, increased generation of hedonic judgments (788). 2) GLP-1 signaling
active GLP-1 in portal-vein plasma to ⬃20-fold the maxi- in the caudal brain stem (624) and in the amygdala (388)
mum level observed after a meal under the same conditions may be involved in the aversive control of eating in rats. 3)
(599, 639). 4) Intraperitoneal injections of an GLP-1-albu- GLP-1 may have physiological roles in thirst and in sodium
min conjugate, which is unlikely to enter the brain, inhib- and water homeostasis (298, 485), which may influence
ited eating in mice (48). 5) In chow-fed rats, intravenous eating under some conditions. Future research should care-
infusions of GLP-1R antagonists failed to increase eating fully consider these possibilities.
(387, 638, 832), whereas intraperitoneal injections of
GLP-1R antagonists did so in several (37, 828, 831), al- There are two interesting distinctions between the eating-
though not all (3, 179, 832), tests. The inconsistent eating- inhibitory effects of chronic GLP-1 treatments in animals
stimulatory effect of intraperitoneal GLP-1-antagonist ad- versus in humans. 1) The site of action in humans is un-
ministration suggests that locally acting GLP-1 may be an known. Chronic GLP-1-agonist treatments that produce
endogenous satiation signal in rats only under particular weight loss in rats do so by acting centrally (379, 677, 698),
conditions. Dietary fat content may be one factor that whereas the long-lasting GLP-1 agonist liraglutide had low
downregulates the eating-inhibitory effect of GLP-1 (224, uptake into the cerebrospinal fluid in humans (150). 2)
595, 831). Visceral illness is not a serious side effect of GLP-1-agonist
treatment in humans (335, 414, 581), but reliably accom- cretin effect was diminished (47, 128, 223, 472). This ap-
panies the reductions in food intake and body weight pro- parently reflects a decrease in the -cell response to GLP-1
duced by chronic GLP-1-agonist treatment in rats (379). and, more importantly, a near lack of response to GIP (332,
GLP-1 agonist-induced illness in rats is apparently medi- 392, 476, 491, 493, 538, 764, 842). As a result, GLP-1 may
ated by a subset of central GLP-1R sites (378). Better un- contribute relatively more to the incretin effect in T2DM
derstanding of these phenomena is important for advancing patients than in metabolically healthy persons (835). The
GLP-1 obesity pharmacotherapy. defect in the incretin effect in T2DM is reversible: improv-
ing glucose levels in patients with T2DM for only 4 wk
D. Glycemic Control improved C-peptide secretion in response to both GLP-1
and GIP infusions markedly (332). Thus GLP-1 agonists
GLP-1 appears to contribute to meal-related glycemic con- hold great promise for the treatment of T2DM. GLP-1-
trol by stimulating insulin secretion, inhibiting glucagon based therapy, however, may not be advantageous for all
secretion, slowing gastric emptying, and reducing hepatic T2DM patients. For example, the discovery of gene poly-
glucose metabolism (128, 222, 336, 653). GLP-1 may also morphisms that affect incretin hormone secretion and ac-
contribute to glycemic control in the fasting state. The latter tion in T2DM patients (527) and that affect GLP-1’s insuli-
is suggested by recent studies employing pancreatic clamps, notropic effect in healthy individuals (657) suggest that de-
i.e., somatostatin infusion and glucagon and insulin re- fects in incretin function may be a primary pathophysiology
placement, that suggest that GLP-1 reduces endogenous in some patients.
glucose production during the fasting state in both metabol-
ically healthy individuals (594) and those with T2DM GLP-1’s glucagonostatic action also contributes to meal-
(679). Studies in mice suggest that these effects are due in
related glycemic control. Physiological infusions of 0.25–
part to an insulin-independent effect GLP-1R in the hepatic
0.4 pmol·kg⫺1·min⫺1 GLP-1 inhibited glucagon secretion
portal vein or liver (117).
and reduced glucose levels in both healthy subjects and
T2DM patients (309, 352), and exendin(9 –39) markedly
GLP-1, together with GIP, mediates the incretin effect by
elevated glucagon secretion and increased glucose levels in
exerting dose-related, glucose-dependent insulinotropic ef-
healthy subjects (230, 546, 721).
fects on -cells (128, 223, 336, 406). Infusions of physio-
logical endocrine doses of GLP-1 are sufficient to increase
As described in section IIC, GLP-1 slows gastric emptying,
insulin secretion in fasting subjects and during glucose in-
which also improves glycemic control (448, 541, 659, 834).
fusions (406, 537, 803). Furthermore, a physiological dose
Comparisons of the relative contributions of GLP-1’s di-
of GLP-1 infused during isoglycemic glucose infusions, i.e.,
infusions leading to identical glycemic profiles as oral glu- verse effects on glycemia suggest that its glucagonostatic
cose, reproduced the insulin response to oral glucose (537). and gastric-emptying inhibitory effects are more important
These data indicate that GLP-1 meets criterion 3 of TABLE 2 than its insulinotropic effect in healthy subjects (541, 546).
for a physiological endocrine incretin effect. Infusion of the Several aspects of GLP-1’s effect on GI motility are relevant
GLP-1R antagonist exendin(9 –39) decreased insulin secre- for the treatment of T2DM. 1) The slowing of gastric emp-
tion after oral glucose loads, after meals, during intraduo- tying by exogenous GLP-1 displayed tachyphylaxis during
denal glucose infusions, and during hyperglycemic glucose sustained (⬎24 h) GLP-1 infusions in healthy subjects (540,
clamps (546, 650, 664, 721), indicating that GLP-1 meets 781). 2) Shorter-acting GLP-1 agonists have a more sus-
also criterion 6 for a physiological endocrine incretin effect. tained effect on gastric emptying and thereby reduce meal-
related glycemia more than longer-acting GLP-1 agonists
Additional important aspects of the incretin effect in meta- (577). 3) A short-acting GLP-1 agonist also reduced duode-
bolically healthy individuals include 1) comparisons of in- nal motility and flow, suggesting an additional mechanism
sulin or C-peptide secretion in response to oral glucose or by which GLP-1 may reduce meal-related glycemia (756).
isoglycemic intravenous glucose infusions indicate that the
incretin effect increases with increasing glucose loads and, Additional mechanisms also may contribute to GLP-1’s gly-
thus, limits meal-related glucose excursions even after large cemic effects. A study in truncally vagotomized human sub-
glucose loads (47, 490, 539). 2) GLP-1 synergizes with GIP jects indicated that the vagus is involved in GLP-1’s effect
to increase insulin secretion (537, 803), but 3) intraduode- on GI glucose disposal, but not in its incretin effect, al-
nal glucose infusions at rates in the physiological range of though a limitation was that the subjects had pyloroplasty
gastric emptying of glucose solutions indicated that GIP together with vagotomy (587). This effect of GLP-1 on
was the predominant incretin during infusion of ⱕ2 kcal/ glucose disposal may involve GLP-1R in the hepatic-portal
min glucose, whereas GLP-1 predominated during infusion vein, which animal studies indicate activate neural reflexes
of either 3 or 4 kcal/min glucose (471, 774). that increase glucose clearance without affecting insulin se-
cretion (117, 355, 548). The role of these reflexes in normal
Although glucose stimulated GLP-1 and GIP secretion nor- meal-related glucose control remains unclear. Mice with
mally in patients with T2DM (47, 121, 460, 542), the in- global deletion of GLP-1R in which GLP-1R were selec-
sham-operated rats, suggesting that central GLP-1R are maturation. In any case, these data provide an important
not involved in the effects of RYGB (848). Unfortunately, challenge to the human literature.
the effects of peripheral GLP-1 were not assessed, so it
was possible that peripheral GLP-1 signaling contributed RYGB increases meal-stimulated circulating bile-acid levels
to the observed effects of RYGB. 4) RYGB had compa- (157, 570, 740), which might contribute to RYGB-induced
rable weight-loss and eating-inhibitory effects in mice increases in GLP-1 secretion and to RYGB’s therapeutic
with transgenic deletions of GLP-1R (Glp1r⫺/⫺) or effects. Consistent with this hypothesis, meal-stimulated
Gnat3 (␣-Gust⫺/⫺), which do not secrete GLP-1 in re- bile-acid and GLP-1 responses were associated in several
sponse to oral glucose, and wild-type mice (507, 848). studies done 4 mo or more after RYGB (396, 566, 668,
Thus the preponderance of evidence from animal models 820). But meal-stimulated bile-acid levels did not increase
argues strongly against the hypothesis that GLP-1 con- in tests 1 wk, 1 mo, or 3 mo after RYGB (16, 720). Thus,
tributes importantly to the eating-inhibitory and weight- because meal-related GLP-1 levels are markedly increased
loss effects of RYGB (the Glp1r⫺/⫺ and ␣-Gust⫺/⫺ mod- at these times, any contribution of elevated bile-acid levels
els are discussed further below). to GLP-1 secretion or the effects of RYGB are likely to be
late-developing mechanisms. Finally, mouse models sup-
In marked contrast to the eating data, there is compelling port the hypothesis that changes in bile acids contribute to
evidence that GLP-1 contributes in several ways to the ben- the effects of bariatric surgery. 1) Diversion of bile to the
eficial effects of RYGB on glycemic regulation in humans. ileum increased circulating bile acids 10-fold and led to
The clearest evidence is that exendin(9 –39) markedly re- decreases in food intake, glycemia, and body weight that
duced insulinemia and increased glycemia after consump- were similar to those produced by RYGB and appeared to
tion of mixed-nutrient meals or glucose solutions tested 1 be at least in part independent of fat malabsorption; unfor-
wk to 5 yr after RYGB (371, 375, 648 – 650, 684, 736). This tunately, the role of GLP-1 was not assessed (257). 2) A
also occurred in RYGB patients with T2DM (371, 375). transgenic mouse-model study (642) implicated FXR in the
Additionally, exendin(9 –39) increased glucagon secretion efficacy of vertical-sleeve gastrectomy; again, the impor-
(375, 650, 736) and accelerated gastric emptying rate in one tance of GLP-1 was not assessed. Furthermore, although
vertical-sleeve gastrectomy increased circulating bile acids
study (684), although not another (650). In contrast to
in mice (529), the human data are mixed (157).
these increased effects of GLP-1, RYGB did not appear to
increase the contribution of GIP to meal-related glycemic
control (736). A dual-isotope glucose-tracing study indi- G. Summary
cated that GLP-1 was not involved in the reduction of en-
dogenous glucose production and the increase in glucose GLP-1 is secreted in response to the products of carbohy-
disposal after RYGB (375). Increased GLP-1 secretion after drate, lipid, and protein digestion. It may act as an endo-
RYGB also contributed to the development of meal-related crine satiation signal in healthy humans, but antagonist
hyperinsulinemic hypoglycemia in some patients (647– 649; studies have not yet confirmed this. Intestinal and brain
for reviews of these and related data, see Refs. 468, 646, GLP-1 also may have other effects on ingestive behavior,
653). It is also important to note that reduced eating also but these are not established in humans. Intestinal GLP-1
contributes importantly to the improvements in glycemic reduces meal-related increases in glycemia by stimulating
control after RYGB (361, 363, 415). insulin secretion (i.e., acting as an incretin), by inhibiting
glucagon secretion, by slowing gastric emptying, and, per-
Exendin(9 –39) treatment also reversed the improvements haps, other effects. The effects of GLP-1 on eating and
in glucose tolerance and insulin secretion after RYGB in a glycemic control are summarized in FIGURE 10. Defects in
rat model (138). There is a disconnect, however, between GLP-1 secretion or signaling may contribute to overeating
the demonstrations with exendin(9 –39) of the importance in obesity. Although GLP-1’s effectiveness in glycemic con-
of GLP-1 for glycemic regulation after RYGB and a report trol decreases in individuals with insulin resistance or
(507) that Glp1r⫺/⫺ and ␣-Gust⫺/⫺ mice with RYGB and T2DM, it remains a crucial contributor, and GLP-1 ago-
wild-type RYGB mice had similar glucose tolerance, insulin nists are already in use in T2DM and obesity therapy.
tolerance, and glucose-stimulated insulin release [␣- GLP-1 contributes to improved glycemic control after
Gust⫺/⫺ mice, which do not secrete measureable GLP-1, RYGB; its role in eating after RYGB is unclear.
were used to test the role of GLP-1’s degradation products
GLP-1(9 –36)amide and GLP-1(28 –36)amide, which may
VI. PYY(3–36)
improve glucose homeostasis via GLP-1R-independent
mechanisms]. The resolution of this apparent paradox is
unclear. It is possible that there are important species dif- A. Introduction
ferences. In addition, because these were germline trans-
genic animals, mechanisms compensating for the lack of Endocrine PYY is synthesized and secreted by open-type
GLP-1 signaling may have developed during the animals’ enteroendocrine cells (FIGURE 6B). Enteroendocrine PYY
ceptors discussed below). Carbohydrate (glucose in most PYY to ⬃45– 60 pM, more than the ⬃40 pM produced by
studies) appears to stimulate PYY secretion in part via stim- a 3000 kcal meal in the same study (423). 3) Infusion of
ulation of TAS1R1/TAS1R3 sweet receptors (228, 275, lower PYY(3–36) doses, 0.2– 0.3 pmol·kg⫺1·min⫺1, failed
717), but because equally sweet artificial sweeteners did not to affect subsequent meal size (194, 701). Thus doses of
trigger PYY release, other mechanisms must be also in- PYY(3–36) that inhibited eating in these studies increased
volved (461, 716, 732, 849). Whether FFAR1 or FFAR4 plasma PYY levels more than all but extremely large meals,
contributes to PYY secretion has not been studied to our suggesting that the effects of PYY(3–36) on postprandial
knowledge, but given that many enteroendocrine cells pro- satiety do not meet criterion 3 of TABLE 2 for a physiological
duce both GLP-1 and PYY (304), it is likely that they do so. endocrine dose. This conclusion is consistent with the re-
CASR appears to contribute to the stimulation of PYY se- port (276) that the 3 h total PYY AUC after high-carbohy-
cretion by oligopeptides and amino acids because their ef- drate, low-fat breakfasts or low-carbohydrate, high-fat
fects on PYY secretion in isolated loops of rat small intes- breakfasts were not significantly correlated with the sizes of
tine were reduced by a CASR inhibitor and dependent on the following lunches, even though ghrelin and GLP-1 AUC
extracellular Ca2⫹ (461). after each breakfast were significantly correlated with lunch
sizes. In addition, although adverse effects were not re-
Neurohumoral reflexes also appear to contribute to PYY ported when 0.8 pmol·kg⫺1·min⫺1 PYY(3–36) was infused
release, especially its early phase (51, 227, 586). 1) Intrave- following standard meals (62, 194, 423), when infused in
nous CCK infusions increased plasma PYY in humans fasting subjects, this dose elicited “severe malaise or nau-
(102). 2) GLP-1 infusion decreased (104) and exendin(9 – sea” in half the subjects in two studies (701, 768). These
39) infusion increased (230, 721) PYY secretion, perhaps data suggest that criterion 5 of TABLE 2, that the effect of
reflecting an autoregulatory mechanism in GLP-1/PYY PYY(3–36) on satiety effect be selective, requires further
cells. 3) Bile acids may be an important mediator, particu- testing. In addition, in all these studies the test meals were
larly of lipid-stimulated PYY secretion (9, 11, 569, 840). 4) offered at fixed times; thus future tests in which subjects are
Animal studies implicated vasoactive intestinal polypeptide asked to report when they wish to initiate meals may reveal
and the vagus nerve in PYY secretion (9, 51–53, 264, 685).
effects of PYY(3–36) on the duration of the intermeal inter-
5) Some mouse PYY cells express Mc4r, which appears to
val, which is hypothesized to be under the control of post-
facilitate PYY secretion (561).
prandial satiety processes (see sect. IB1).
(517). In one study, hepatic-portal infusions of PYY(3–36) more, as described in section IB2, some PYY is apparently
inhibited eating in rats without signs of illness (710), but released from neuropods (92, 93), and analysis of the po-
systemic intravenous infusions of PYY(3–36) that inhibited tential effects of this is beyond available physiological meth-
eating in rats did produce illness (143, 144), as did intra- ods. Finally, Batterham and colleagues (64, 467) hypothe-
peritoneal injections of PYY(3–36) in mice (306). Interest- sized that PYY is involved in protein-induced satiety and in
ingly, however, intravenous infusions of a peripherally act- exercise-induced anorexia, which have not been extensively
ing NPY2R antagonist reduced the eating-inhibitory effects tested, and in food reward, which has been tested in a num-
of intravenous infusions of PYY(3–36) and of smaller, but ber of human functional brain imaging studies that we do
not larger, intragastric loads of protein and fat in rats (614), not review (63, 194, 818).
suggesting that PYY(3–36) fulfills criterion 6 of TABLE 2
under some conditions. Infusion of the NPY2R antagonist
by itself failed to increase eating, however, which fails to
D. Glycemic Control
provide support for criterion 6 of TABLE 2. Studies of mice
with transgenic deletions of Pyy also provide only weak There is little evidence that PYY(3–36) affects insulin secre-
support for a role in eating (441). In one Pyy⫺/⫺ mouse line, tion in humans. Infusion of 1 or 5 pmol·kg⫺1·min⫺1
male mice were hyperphagic and females were not tested PYY(3–36) failed to affect the insulin response to a bolus
(64); in another, females but not males were hyperphagic intravenous glucose infusion in fasting women (19), and
(88); and in two others no eating phenotype was detected PYY(3–36) infusions during meals failed to increase plasma
(669, 838). Ectopic Pyy overexpression in adult mice failed insulin levels (61, 62), except when the doses elicited illness
(701, 834).
to affect body weight, but slightly decreased eating after a
24 h fast (686). As none of these transgenic methods dis-
In contrast, animal studies suggest that PYY affects glycemic
criminated between effects of PYY(1–36) and PYY(3–36),
regulation in two apparently opposing ways. 1) PYY(3–36)
it is possible that more refined molecular genetic tools will
may indirectly stimulate nutrient-induced insulin secretion in
provide more useful information. Finally, although knock-
rodents. PYY(3–36) reduced postprandial glycemia without
out of peripheral NPY2R in mice increased eating under
affecting fasting glycemia, an effect mimicked by a NPY2R
some conditions, it appeared that this was secondary to
agonist, and this was blocked by peripheral, but not central,
metabolic effects (687, 851).
administration of a NPY2R antagonist (140, 467). This effect
of PYY(3–36) appeared to be mediated by GLP-1 because
Whether PYY(3–36) acts peripherally or centrally to inhibit
exendin(9 –39) blocked the glucose-lowering effects of
eating is unclear. In support of peripheral action, 1) subdi-
PYY(3–36) (140, 467). 2) PYY(1–36) may directly inhibit in-
aphragmatic vagotomy reduced or abolished the eating- sulin secretion via a paracrine mode of action. In mice, PYY is
inhibitory effect of peripherally administered PYY(3–36) in expressed in pancreatic ␣- and ␦-cells, Npy1r and Npy4r, but
rats (56, 395), 2) conjugating PYY(3–36) to albumin to not Npy2r, are expressed in -cells, and PYY(1–36), but not
prevent it from crossing the blood-brain barrier reduced its PYY(3–36), dose-dependently reduced glucose-stimulated in-
eating-inhibitory potency (56), and 3) an NPY2R antago- sulin release from -cells in vitro (116, 140, 467, 573, 651,
nist that does not cross the blood-brain barrier blocked 790). Furthermore, this was absent in cells derived from
the eating-inhibitory effect of peripherally administered Pyy⫺⫺/⫺ or Npy1r⫺/⫺ mice, and both mutants were hyperin-
PYY(3–36) (614). In support of central action, 1) injections sulinemic (88, 116).
of PYY(3–36) directly into the Arc reduced eating in rats
(2), 2) injections of an NPY2R antagonist into the Arc re- PYY may contribute to glycemic regulation via two further
duced the eating-inhibitory effect of peripherally adminis- actions. 1) Endocrine intestinal PYY(3–36) may improve in-
tered PYY(3–36) in rats (2), 3) PYY(3–36) inhibited eating sulin sensitivity, at least under some conditions, because intra-
in vagotomized mice (306), and 4) PYY(3–36) inhibited venous infusion of PYY(3–36) increased glucose uptake in
eating in rats with capsaicin lesions of unmyelinated ab- muscle and adipose tissue of high-fat fed mice during a hyper-
dominal afferents (613). insulinemic-euglycemic clamp (790). 2) Paracrine pancreatic
PYY(1–36) may tonically stimulate islet-cell proliferation and
In conclusion, present data fail to support the hypothesis inhibit -cell apoptosis in mice (573, 651). 3) Any decrease in
that PYY(3–36) physiologically inhibits eating in humans gastric emptying produced by PYY(3–36) may lead to reduc-
or animals. Further efforts to determine whether PYY(3– tions in glycemia.
36) infusions that better model the dynamics of human and
animal PYY secretion around meals have physiological eat- Interestingly, oral fat loads and mixed-nutrient meals ap-
ing-inhibitory effects are required to determine whether pear to stimulate less PYY secretion in patients with T2DM
PYY(3–36) is a plausible candidate physiological satiation (238, 252, 856). To investigate whether this precedes
or postprandial satiety signal. Studies of antagonism of T2DM, Viardot et al. (801) compared subjects with strong
PYY(3–36)-NPY2R signaling are also necessary, but spe- family histories of T2DM with subjects matched for insulin
cific antagonists for human use are not available. Further- sensitivity, age, and BMI, but without family histories of
T2DM. PYY responses to high-carbohydrate meals were different if tested when RYGB reduced eating. 2) Pyy⫺/⫺
impaired in the subjects at risk for T2DM, suggesting that mice lost less weight than wild-type mice during the initial
defective PYY secretion may be causally linked with 10 days after surgical bypass of the duodenum and proxi-
T2DM. mal jejunum; unfortunately, food intakes were not reported
(141). Finally, as noted in section VIF, increases in PYY(3–
36) secretion in patients bearing the MC4R variant I251L
E. Obesity (469) could explain their better weight-loss outcomes after
RYGB (502).
The relationship between obesity and PYY secretion is un-
clear. 1) Although several studies detected decreases in fast-
ing total PYY levels in obese patients (61, 64, 423, 633, G. Summary
856), other similarly powered studies did not (333, 385,
576, 727, 794). 2) Weight reduction was reported to in- PYY(3–36) is secreted in response to the products of carbo-
crease fasting total PYY in obese children (633), but to hydrate, lipid, and protein digestion during and after meals.
decrease it in obese adults (576), although in both studies As summarized in FIGURE 11, PYY(3–36) may contribute to
the weight and PYY changes were small. 3) Postprandial
PYY secretion was reduced in obese patients in six studies
(61, 64, 422, 423, 497, 727, 856), but not in four others PYY(3-36) 1 Satiation
(103, 333, 385, 794). 4) In the one comparison of PYY(3–
36)’s eating-inhibitory effect in obese and healthy-weight
subjects to date, no difference was detected (61).
F. RYGB
gastric emptying via the ileal brake mechanism, to the inhi- won is not a criticism of the strategy. Rather, criteria for
bition of eating, and to the control of meal-related glycemia, physiological function should continue to guide GI hor-
but the evidence that these are physiological actions re- mone research. Identifying truly physiological endocrine
mains thin. Similarly, PYY(3–36)’s role in RYGB remains functions of GI hormones can only facilitate understanding
unclear. This modest progress may be due in part to the of eating, GI motor function, and meal-related glycemic
difficulties of PYY(3–36) research, including the low control and development of therapies for their disorders.
threshold for eliciting illness with PYY(3–36) infusions, the
lack of NPY2R antagonists for human use, and the possi- Perhaps the most pressing issue facing ghrelin, CCK,
bility of neuropod PYY signaling. GLP-1, and PYY(3–36) physiology is the need to determine
the roles of non-endocrine, i.e., local, signaling, which has
been implicated in several of the effects reviewed. The need
VII. DISCUSSION to develop methods enabling tests of local-signaling hy-
potheses against the criteria of TABLE 2 is especially urgent.
The act of eating sets in motion an intricately coordinated As mentioned in section IVD, intraintestinal hormone infu-
series of GI responses that, via central and peripheral influ- sions might selectively target the lamina propria (147), and
ences, contribute importantly to the control of eating and hormone concentrations in the lamina propria can be esti-
meal-related glycemia. The control of secretion of GI hor- mated from assays of lymph. The temporal resolution of
mones by small-intestinal nutrient sensing is a cornerstone lymph assays, however, is poor due to its slow flow. Nor
of these functions. The hormones control exposure of the have the results of tests of meal-related hormone changes in
small intestine to nutrients via their effects on GI motility, the lymph been straightforward. For example, post-meal
especially gastric emptying, and thereby modulate their concentrations of GLP-1 were reported to be ⬃6-fold
own secretion. Here we reviewed the nutrient-secretory higher in lymph than in hepatic portal-vein plasma in rats
controls and contributions to eating, meal-related glycemia, (178) and ⬃8-fold higher in lymph than in orbital-plexus
and GI motility of ghrelin, CCK, GLP-1, and PYY(3–36), in plasma in mice (553), but ⬃10-fold lower in lymph in he-
healthy-weight and obese humans as well as in RYGB pa- patic portal-vein plasma in swine (308).
tients. The primary focus was on normal endogenous or
“physiological” endocrine function in humans because cur- An additional, related challenge for GI-hormonal physiol-
rently available methods make its determination feasible ogy is to encompass the emerging picture of integrated hor-
(TABLE 2). monal and electric signaling in the GI tract. Electrically
excitable GI cells form what Bohórquez and Liddle (91) call
As TABLE 5 indicates, despite considerable research effort, the gut connectome, comprised of enteroendocrine cells,
at present there are many more questions than answers neurons and glia of the enteric nervous system, intrinsic GI
regarding ghrelin, CCK, GLP-1, and PYY(3–36) as physio- neurons, and peripheral ganglia innervating the GI tract.
logical endocrine signals in humans in the functions reviewed. Finally, a third challenge is to better link GI-hormonal phys-
Indeed, only CCK has been fully established as a physiological iology to the study of information processing in the brain,
endocrine control of eating and only GLP-1 as a physiological
endocrine control of meal-related glycemia in healthy-weight
individuals. There is incomplete support for endocrine roles of
CCK in meal-related glycemia and gastric emptying, for Table 5. Physiological status of ghrelin, CCK, GLP-1, and
PYY(3–36) in the endocrine control of eating, GI motility, and
GLP-1 in eating and gastric emptying, and for PYY(3–36) in
meal-related glycemic control in healthy humans
gastric emptying in humans. Moreover, animal research fills
only a few of the gaps indicated in TABLE 5 (an important Meal-Related
exception is that GLP-1R antagonism does increase eating in Eating GI Motility* Glycemic Control
rats under many conditions; see sect. VD). PD/A PD/A PD/A
In view of this decidedly modest estimation of the state of Ghrelin ?/? ?/? ?/?
proof of physiological endocrine function for ghrelin, CCK, CCK YES/YES Yes/Yes† YES/No
GLP-1, and PYY(3–36), one may question whether the cri- GLP-1 YES/No‡ Yes/Yes YES/YES
teria for physiological function (TABLE 2) are overly rigor- PYY(3–36) No/? Yes/? ?/?
ous or whether the criterion-based approach is misguided. Experimental support for the two cardinal criteria of physiological
The answer to each question is no. As reflected in TABLE 1, endocrine function, i.e., the “physiological-dose” criterion (PD) and
criteria for endocrine function have evolved in step with “antagonism” criterion (A), is rated as convincing (YES), partial (Yes),
negative (No), or unknown (?) for each hormone and function. See
advances in understanding and methodology during the text for details and references. *GI motility refers to gastric emptying
century-plus history of endocrinology and have shaped the and small-intestinal motor function. †Cholecystokinin (CCK) antago-
logical and programmatic course of endocrinology and its nism slowed gastric emptying of liquid food, but not solid food. ‡In
two studies (489, 714), premeal administration of the GLP-1 recep-
contributions to medical diagnosis and treatment (59, 292, tor antagonist exendin-9 failed to increase eating, although in one
449, 489, 610, 833). That knowledge at each stage is hard study (714) subjective ratings of appetite increased.
as understood using functional imaging methods in humans Unfamiliar Food Same Food,
and neuropharmacological and molecular-genetic methods Now Familiar
in animals. Present progress in humans is limited largely to -20%
studies of the telencephalic mechanisms of food hedonics, -40%
which have been reviewed elsewhere (references are given in
sect. IA). Advances in the spatial resolution of functional
imaging methods should allow this kind of work to address
the diencephalic and brain-stem mechanisms that are
clearly crucial for the effects of GI hormones on other as-
pects of eating as well as for the control of GI motility and
metabolic function. Increased back-and-forth translation Saline CCK Saline CCK
between human and animal studies should also accelerate FIGURE 12. A thought experiment depicting how learning may
progress in unraveling central mechanisms mediating the influence the control of eating by GI hormones. Left: when an indi-
vidual is served a palatable but unfamiliar food, meal size is deter-
functions of ghrelin, CCK, GLP-1, and PYY(3–36).
mined mainly by unconditioned satiation signals related to gastric
volume, CCK and GLP-1 secretion, etc., as discussed in the review.
Future research should also widen the range of designs used in Under these conditions, CCK infusion during the meal might exert its
GI hormone physiology. Studies of meal size and timing in full unconditioned effect, indicated by the 40% reduction in meal
particular have used remarkably similar experimental ap- size. Right: if the same individual is tested after extensive experience
eating the test food, meal size might be the same as initially, but will
proaches, which may contribute to some of the negative data now be under the control of conditioned responses such as expected
reviewed. As discussed in section IB2, few GI hormone-infu- satiation (111), portion-size estimation (629), etc., that override
sion studies have interrogated parameters other than peak unconditioned signals, and because conditioned eating controls are
plasma levels. Studies of rate ascent of plasma hormone con- resistant to physiological feedback, the same CCK infusion might
centration, the timing of the infusion in relation to the course now reduce meal size less, indicated by the 20% effect.
hormones. Although there are presently few methods to 5. Acosta A, Camilleri M, Shin A, Vazquez-Roque MI, Iturrino J, Burton D, O’Neill J,
study the physiology of such effects, emerging technolo- Eckert D, Zinsmeister AR. Quantitative gastrointestinal and psychological traits asso-
ciated with obesity and response to weight-loss therapy. Gastroenterology 148: 537–
gies for miniaturization, telemetry, and molecular-ge-
546, 2015.
netic methods applicable to humans (209, 416, 428, 559,
816) may soon create new opportunities. These, as well 6. Adam TC, Westerterp-Plantenga MS. Glucagon-like peptide-1 release and satiety
after a nutrient challenge in normal-weight and obese subjects. Br J Nutr 93: 845– 851,
as more sophisticated testing designs, should be exploited 2005.
to expand basic physiological knowledge and to help
meet the continuing challenges of the epidemics of obe- 7. Adolph EF. Early concepts of physiological regulations. Physiol Rev 41: 737–770, 1961.
sity and T2DM. 8. Adrian TE, Bacarese-Hamilton AJ, Smith HA, Chohan P, Manolas KJ, Bloom SR.
Distribution and postprandial release of porcine peptide YY. J Endocrinol 113: 11–14,
1987.
ACKNOWLEDGMENTS
9. Adrian TE, Ballantyne GH, Longo WE, Bilchik AJ, Graham S, Basson MD, Tierney RP,
We thank Timothy H. Moran, Johns Hopkins University Modlin IM. Deoxycholate is an important releaser of peptide YY and enteroglucagon
from the human colon. Gut 34: 1219 –1224, 1993.
School of Medicine; Gerard P. Smith, Weill Medical Col-
lege of Cornell University; Stephen C. Woods, University 10. Adrian TE, Ferri GL, Bacarese-Hamilton AJ, Fuessl HS, Polak JM, Bloom SR. Human
distribution and release of a putative new gut hormone, peptide YY. Gastroenterology
of Cincinnati; Guillaume de Lartigue, John B. Pierce Lab-
89: 1070 –1077, 1985.
oratory; and Richard L. Young and Tongzhi Wu, Disci-
pline of Medicine, The University of Adelaide, for helpful 11. Adrian TE, Gariballa S, Parekh KA, Thomas SA, Saadi H, Al Kaabi J, Nagelkerke N,
Gedulin B, Young AA. Rectal taurocholate increases L cell and insulin secretion, and
comments. decreases blood glucose and food intake in obese type 2 diabetic volunteers. Diabe-
tologia 55: 2343–2347, 2012.
Address for reprint requests and other correspondence:
12. Adrian TE, Long RG, Fuessl HS, Bloom SR. Plasma peptide YY (PYY) in dumping
R. E. Steinert, DSM Nutritional Products Ltd., R&D Hu- syndrome. Dig Dis Sci 30: 1145–1148, 1985.
man Nutrition and Health, Basel, Switzerland (e-mail: re.
[email protected]). 13. Adrian TE, Savage AP, Bacarese-Hamilton AJ, Wolfe K, Besterman HS, Bloom SR.
Peptide YY abnormalities in gastrointestinal diseases. Gastroenterology 90: 379 –384,
1986.
GRANTS
14. Adrian TE, Savage AP, Fuessl HS, Wolfe K, Besterman HS, Bloom SR. Release of
peptide YY (PYY) after resection of small bowel, colon, or pancreas in man. Surgery
R. E. Steinert was supported by a Mary Overton Early 101: 715–719, 1987.
Career Fellowship, C. Feinle-Bisset by a Senior Research
15. Ahlkvist L, Vikman J, Pacini G, Ahren B. Synergism by individual macronutrients ex-
Fellowship from the National Health & Medical Research plains the marked early GLP-1 and islet hormone responses to mixed meal challenge
Council of Australia (Grant 627002), L. Asarian by U.S. in mice. Regul Pept 178: 29 –35, 2012.
National Institutes of Health Grant DK-092608, and C.
16. Ahmad NN, Pfalzer A, Kaplan LM. Roux-en-Y gastric bypass normalizes the blunted
Beglinger by Swiss National Science Foundation Grant postprandial bile acid excursion associated with obesity. Int J Obes 37: 1553–1559,
320030-132960/1. 2013.
17. Ahrén B, Holst JJ. The cephalic insulin response to meal ingestion in humans is depen- 36. Asakawa A, Inui A, Kaga T, Yuzuriha H, Nagata T, Ueno N, Makino S, Fujimiya M,
dent on both cholinergic and noncholinergic mechanisms and is important for post- Niijima A, Fujino MA, Kasuga M. Ghrelin is an appetite-stimulatory signal from stom-
prandial glycemia. Diabetes 50: 1030 –1038, 2001. ach with structural resemblance to motilin. Gastroenterology 120: 337–345, 2001.
18. Ahren B, Holst JJ, Efendic S. Antidiabetogenic action of cholecystokinin-8 in type 2 37. Asarian L, Abegg K, Geary N, Schiesser M, Lutz TA, Bueter M. Estradiol increases
diabetes. J Clin Endocrinol Metab 85: 1043–1048, 2000. body weight loss and gut-peptide satiation after Roux-en-Y gastric bypass in ovariec-
tomized rats. Gastroenterology 143: 325–327, 2012.
19. Ahren B, Larsson H. Peptide YY does not inhibit glucose-stimulated insulin secretion
in humans. Eur J Endocrinol 134: 362–365, 1996. 38. Asarian L, Geary N. Sex differences in the physiology of eating. Am J Physiol Regul Integr
Comp Physiol 305: R1215–R1267, 2013.
20. Akamizu T, Takaya K, Irako T, Hosoda H, Teramukai S, Matsuyama A, Tada H, Miura
K, Shimizu A, Fukushima M, Yokode M, Tanaka K, Kangawa K. Pharmacokinetics, 39. Ashrafian H, le Roux CW. Metabolic surgery and gut hormones: a review of bariatric
safety, and endocrine and appetite effects of ghrelin administration in young healthy entero-humoral modulation. Physiol Behav 97: 620 – 631, 2009.
subjects. Eur J Endocrinol 150: 447– 455, 2004.
40. Atalayer D, Astbury NM. Anorexia of aging and gut hormones. Aging Dis 4: 264 –275,
21. Akkary E, Sidani S, Boonsiri J, Yu S, Dziura J, Duffy AJ, Bell RL. The paradox of the 2013.
pouch: prompt emptying predicts improved weight loss after laparoscopic Roux-Y
gastric bypass. Surg Endosc 23: 790 –794, 2009. 41. Aulinger BA, Bedorf A, Kutscherauer G, de Heer J, Holst JJ, Goke B, Schirra J. Defining
the role of GLP-1 in the enteroinsulinar axis in type 2 diabetes (T2D) utilizing DPP-4
22. Al Massadi O, Pardo M, Roca-Rivada A, Castelao C, Casanueva FF, Seoane LM. inhibition and GLP-1-receptor blockade. Diabetes 63: 1079 –1092, 2014.
Macronutrients act directly on the stomach to regulate gastric ghrelin release. J En-
docrinol Invest 33: 599 – 602, 2010. 42. Aulinger BA, Vahl TP, Wilson-Perez HE, Prigeon RL, D’Alessio DA. -Cell sensitivity
to GLP-1 in healthy humans is variable and proportional to insulin sensitivity. J Clin
23. Alhadeff AL, Baird JP, Swick JC, Hayes MR, Grill HJ. Glucagon-like Peptide-1 receptor
Endocrinol Metab 100: 2489 –2496, 2015.
signaling in the lateral parabrachial nucleus contributes to the control of food intake
and motivation to feed. Neuropsychopharmacology 39: 2233–2243, 2014. 43. Azpiroz F, Feinle-Bisset C, Grundy D, Tack J. Gastric sensitivity and reflexes: basic
mechanisms underlying clinical problems. J Gastroenterol 49: 206 –218, 2014.
24. Alhadeff AL, Rupprecht LE, Hayes MR. GLP-1 neurons in the nucleus of the solitary
tract project directly to the ventral tegmental area and nucleus accumbens to control 44. Babkin BP. Pavlov—a Biography. Chicago, IL: Univ. of Chicago Press, 1949.
for food intake. Endocrinology 153: 647– 658, 2012.
45. Bacarese-Hamilton AJ, Adrian TE, Bloom SR. Distribution and heterogeneity of im-
25. Ali-Rachedi A, Varndell IM, Adrian TE, Gapp DA, Van Noorden S, Bloom SR, Polak JM. munoreactive cholecystokinin (CCK) in the mucosa of the porcine gastrointestinal
Peptide YY (PYY) immunoreactivity is co-stored with glucagon-related immunoreac- tract. Regul Pept 9: 289 –298, 1984.
tants in endocrine cells of the gut and pancreas. Histochemistry 80: 487– 491, 1984.
46. Bach Knudsen KE, Jorgensen H, Canibe N. Quantification of the absorption of nutri-
26. Allen JM, Fitzpatrick ML, Yeats JC, Darcy K, Adrian TE, Bloom SR. Effects of peptide
ents derived from carbohydrate assimilation: model experiment with catheterised
YY and neuropeptide Y on gastric emptying in man. Digestion 30: 255–262, 1984.
pigs fed on wheat- or oat-based rolls. Br J Nutr 84: 449 – 458, 2000.
27. Alsalim W, Omar B, Pacini G, Bizzotto R, Mari A, Ahren B. Incretin and islet hormone
47. Bagger JI, Knop FK, Lund A, Vestergaard H, Holst JJ, Vilsbøll T. Impaired regulation of
responses to meals of increasing size in healthy subjects. J Clin Endocrinol Metab 100:
the incretin effect in patients with type 2 diabetes. J Clin Endocrinol Metab 96: 737–
561–568, 2015.
745, 2011.
28. Alvarez E, Martinez MD, Roncero I, Chowen JA, Garcia-Cuartero B, Gispert JD, Sanz
48. Baggio LL, Huang Q, Brown TJ, Drucker DJ. A recombinant human glucagon-like
C, Vazquez P, Maldonado A, de Caceres J, Desco M, Pozo MA, Blazquez E. The
peptide (GLP)-1-albumin protein (albugon) mimics peptidergic activation of GLP-1
expression of GLP-1 receptor mRNA and protein allows the effect of GLP-1 on
receptor-dependent pathways coupled with satiety, gastrointestinal motility, and glu-
glucose metabolism in the human hypothalamus and brainstem. J Neurochem 92:
cose homeostasis. Diabetes 53: 2492–2500, 2004.
798 – 806, 2005.
29. Anini Y, Hansotia T, Brubaker PL. Muscarinic receptors control postprandial release 49. Bak MJ, Wewer Albrechtsen NJ, Pedersen J, Knop FK, Vilsboll T, Jorgensen NB,
of glucagon-like peptide-1: in vivo and in vitro studies in rats. Endocrinology 143: Hartmann B, Deacon CF, Dragsted LO, Holst JJ. Specificity and sensitivity of com-
2420 –2426, 2002. mercially available assays for glucagon-like peptide-1 (GLP-1): implications for GLP-1
measurements in clinical studies. Diabetes Obes Metab 16: 1155–1164, 2014.
30. Antoniou AJ, Raja S, El-Khouli R, Mena E, Lodge MA, Wahl RL, Clarke JO, Pasricha P,
Ziessman HA. Comprehensive radionuclide esophagogastrointestinal transit study: 50. Balks HJ, Holst JJ, von zur Muhlen A, Brabant G. Rapid oscillations in plasma glucagon-
methodology, reference values, and initial clinical experience. J Nucl Med 56: 721– like peptide-1 (GLP-1) in humans: cholinergic control of GLP-1 secretion via musca-
727, 2015. rinic receptors. J Clin Endocrinol Metab 82: 786 –790, 1997.
31. Aprahamian CJ, Tekant G, Chen M, Yagmurlu A, Yang YK, Loux T, Harmon CM. A rat 51. Ballantyne GH. Peptide YY(1–36) and peptide YY(3–36): Part I. Distribution, release
model of childhood diet-induced obesity: Roux-en-Y gastric bypass induced changes and actions. Obes Surg 16: 651– 658, 2006.
in metabolic parameters and gastric peptide ghrelin. Pediatr Surg Int 23: 653– 657,
2007. 52. Ballantyne GH, Goldenring JR, Savoca PE, Kranz HK, Adrian TE, Bilchik AJ, Modlin IM.
Cyclic AMP-mediated release of peptide YY (PYY) from the isolated perfused rabbit
32. Ard J, Cannon A, Lewis CE, Lofton H, Vang Skjoth T, Stevenin B, Pi-Sunyer X. Efficacy distal colon. Regul Pept 47: 117–126, 1993.
and safety of liraglutide 30 mg for weight management are similar across races:
subgroup analysis across the SCALE and phase II randomized trials. Diabetes Obes 53. Ballantyne GH, Longo WE, Savoca PE, Adrian TE, Vukasin AP, Bilchik AJ, Sussman J,
Metab 18: 430 – 435, 2016. Modlin IM. Deoxycholate-stimulated release of peptide YY from the isolated perfused
rabbit left colon. Am J Physiol Gastrointest Liver Physiol 257: G715–G724, 1989.
33. Arkwright JW, Blenman NG, Underhill ID, Maunder SA, Spencer NJ, Costa M,
Brookes SJ, Szczesniak MM, Dinning PG. A fibre optic catheter for simultaneous 54. Ballinger A, McLoughlin L, Medbak S, Clark M. Cholecystokinin is a satiety hormone
measurement of longitudinal and circumferential muscular activity in the gastrointes- in humans at physiological post-prandial plasma concentrations. Clin Sci 89: 375–381,
tinal tract. J Biophotonics 4: 244 –251, 2011. 1995.
34. Arnold M, Mura A, Langhans W, Geary N. Gut vagal afferents are not necessary for the 55. Ballinger AB, Clark ML. L-Phenylalanine releases cholecystokinin (CCK) and is asso-
eating-stimulatory effect of intraperitoneally injected ghrelin in the rat. J Neurosci 26: ciated with reduced food intake in humans: evidence for a physiological role of CCK
11052–11060, 2006. in control of eating. Metabolism 43: 735–738, 1994.
35. Arosio M, Ronchi CL, Beck-Peccoz P, Gebbia C, Giavoli C, Cappiello V, Conte D, 56. Baraboi ED, Michel C, Smith P, Thibaudeau K, Ferguson AV, Richard D. Effects of
Peracchi M. Effects of modified sham feeding on ghrelin levels in healthy human albumin-conjugated PYY on food intake: the respective roles of the circumventricular
subjects. J Clin Endocrinol Metab 89: 5101–5104, 2004. organs and vagus nerve. Eur J Neurosci 32: 826 – 839, 2010.
57. Baranowska B, Radzikowska M, Wasilewska-Dziubinska E, Roguski K, Borowiec M. 78. Bhattacharya SK, Andrews K, Beveridge R, Cameron KO, Chen C, Dunn M, Fernando
Disturbed release of gastrointestinal peptides in anorexia nervosa and in obesity. D, Gao H, Hepworth D, Jackson VM, Khot V, Kong J, Kosa RE, Lapham K, Loria PM,
Diabetes Obes Metab 2: 99 –103, 2000. Londregan AT, McClure KF, Orr ST, Patel J, Rose C, Saenz J, Stock IA, Storer G,
VanVolkenburg M, Vrieze D, Wang G, Xiao J, Zhang Y. Discovery of PF-5190457, a
58. Barnett BP, Hwang Y, Taylor MS, Kirchner H, Pfluger PT, Bernard V, Lin YY, Bowers potent, selective, and orally bioavailable ghrelin receptor inverse agonist clinical can-
EM, Mukherjee C, Song WJ, Longo PA, Leahy DJ, Hussain MA, Tschop MH, Boeke JD, didate. ACS Med Chem Lett 5: 474 – 479, 2014.
Cole PA. Glucose and weight control in mice with a designed ghrelin O-acyltrans-
ferase inhibitor. Science 330: 1689 –1692, 2010. 79. Bi S, Scott KA, Kopin AS, Moran TH. Differential roles for cholecystokinin a receptors
in energy balance in rats and mice. Endocrinology 145: 3873–3880, 2004.
59. Barrington EJW. An Introduction to General and Comparative Endocrinology. Oxford,
UK: Clarendon, 1963, p. 33–35. 80. Birch LL, Doub AE. Learning to eat: birth to age 2 y. Am J Clin Nutr 99: 723S–728S,
2014.
60. Bartoshuk LM, Duffy VB, Hayes JE, Moskowitz HR, Snyder DJ. Psychophysics of
sweet and fat perception in obesity: problems, solutions and new perspectives. Philos 81. Bjorklund P, Laurenius A, Een E, Olbers T, Lonroth H, Fandriks L. Is the Roux limb a
Trans R Soc Lond B Biol Sci 361: 1137–1148, 2006. determinant for meal size after gastric bypass surgery? Obes Surg 20: 1408 –1414,
2010.
61. Batterham RL, Cohen MA, Ellis SM, Le Roux CW, Withers DJ, Frost GS, Ghatei MA,
Bloom SR. Inhibition of food intake in obese subjects by peptide YY3-36. N Engl J Med 82. Blackshaw LA, Brookes SJ, Grundy D, Schemann M. Sensory transmission in the
349: 941–948, 2003. gastrointestinal tract. Neurogastroenterol Motil 19: 1–19, 2007.
62. Batterham RL, Cowley MA, Small CJ, Herzog H, Cohen MA, Dakin CL, Wren AM, 83. Blevins JE, Stanley BG, Reidelberger RD. Brain regions where cholecystokinin sup-
Brynes AE, Low MJ, Ghatei MA, Cone RD, Bloom SR. Gut hormone PYY(3–36) presses feeding in rats. Brain Res 860: 1–10, 2000.
physiologically inhibits food intake. Nature 418: 650 – 654, 2002.
84. Blom WA, de Graaf C, Lluch A, Stafleu A, Schaafsma G, Hendriks HF. Postprandial
63. Batterham RL, Ffytche DH, Rosenthal JM, Zelaya FO, Barker GJ, Withers DJ, Williams ghrelin responses are associated with the intermeal interval in time-blinded normal
SCR. PYY modulation of cortical and hypothalamic brain areas predicts feeding be- weight men, but not in obese men. Physiol Behav 96: 742–748, 2009.
haviour in humans. Nature 450: 106 –109, 2007.
85. Blundell J. Pharmacological approaches to appetite suppression. Trends Pharmacol Sci
64. Batterham RL, Heffron H, Kapoor S, Chivers JE, Chandarana K, Herzog H, Le Roux 12: 147–157, 1991.
CW, Thomas EL, Bell JD, Withers DJ. Critical role for peptide YY in protein-mediated
satiation and body-weight regulation. Cell Metab 4: 223–233, 2006. 86. Blundell J, de Graaf C, Hulshof T, Jebb S, Livingstone B, Lluch A, Mela D, Salah S,
Schuring E, van der Knaap H, Westerterp M. Appetite control: methodological as-
65. Baum F, Nauck MA, Ebert R, Cantor P, Hoffmann G, Choudhury AR, Schmidt WE, pects of the evaluation of foods. Obes Rev 11: 251–270, 2010.
Creutzfeldt W. Role of endogenously released cholecystokinin in determining post-
prandial insulin levels in man: effects of loxiglumide, a specific cholecystokinin recep- 87. Boesmans W, Hao MM, Vanden Berghe P. Optical tools to investigate cellular activity
tor antagonist. Digestion 53: 189 –199, 1992. in the intestinal wall. J Neurogastroenterol Motil 21: 337–351, 2015.
66. Bayliss WM, Starling EH. The mechanism of pancreatic secretion. J Physiol 28: 325– 88. Boey D, Lin S, Karl T, Baldock P, Lee N, Enriquez R, Couzens M, Slack K, Dallmann R,
353, 1902. Sainsbury A, Herzog H. Peptide YY ablation in mice leads to the development of
hyperinsulinaemia and obesity. Diabetologia 49: 1360 –1370, 2006.
67. Beardshall K, Frost G, Morarji Y, Domin J, Bloom SR, Calam J. Saturation of fat and
cholecystokinin release: implications for pancreatic carcinogenesis. Lancet 2: 1008 – 89. Bohdjalian A, Ludvik B, Guerci B, Bresler L, Renard E, Nocca D, Karnieli E, Assalia A,
1010, 1989. Prager R, Prager G. Improvement in glycemic control by gastric electrical stimulation
(TANTALUS) in overweight subjects with type 2 diabetes. Surg Endosc 23: 1955–
68. Beauchamp GK, Moran M. Dietary experience and sweet taste preference in human 1960, 2009.
infants. Appetite 3: 139 –152, 1982.
90. Bohorquez DV, Liddle RA. Axon-like basal processes in enteroendocrine cells: char-
69. Beglinger C, Degen L. Gastrointestinal satiety signals in humans—physiologic roles for acteristics and potential targets. Clin Transl Sci 4: 387–391, 2011.
GLP-1 and PYY? Physiol Behav 89: 460 – 464, 2006.
91. Bohorquez DV, Liddle RA. The gut connectome: making sense of what you eat. J Clin
70. Beglinger C, Degen L, Matzinger D, D’Amato M, Drewe J. Loxiglumide, a CCK-A Invest 125: 888 – 890, 2015.
receptor antagonist, stimulates calorie intake and hunger feelings in humans. Am J
Physiol Regul Integr Comp Physiol 280: R1149 –R1154, 2001. 92. Bohorquez DV, Samsa LA, Roholt A, Medicetty S, Chandra R, Liddle RA. An enteroen-
docrine cell-enteric glia connection revealed by 3D electron microscopy. PLoS One 26:
71. Beglinger S, Drewe J, Schirra J, Göke B, D’Amato M, Beglinger C. Role of fat hydrolysis e89881, 2014.
in regulating glucagon-like Peptide-1 secretion. J Clin Endocrinol Metab 95: 879 – 886,
2010. 93. Bohorquez DV, Shahid RA, Erdmann A, Kreger AM, Wang Y, Calakos N, Wang F,
Liddle RA. Neuroepithelial circuit formed by innervation of sensory enteroendocrine
72. Bell GI, Sanchez-Pescador R, Laybourn PJ, Najarian RC. Exon duplication and diver- cells. J Clin Invest 125: 782–786, 2015.
gence in the human preproglucagon gene. Nature 304: 368 –371, 1983.
94. Borg CM, le Roux CW, Ghatei MA, Bloom SR, Patel AG, Aylwin SJ. Progressive rise in
73. Bennink R, Peeters M, Van den Maegdenbergh V, Geypens B, Rutgeerts P, De Roo M, gut hormone levels after Roux-en-Y gastric bypass suggests gut adaptation and ex-
Mortelmans L. Comparison of total and compartmental gastric emptying and antral plains altered satiety. Br J Surg 93: 210 –215, 2006.
motility between healthy men and women. Eur J Nucl Med 25: 1293–1299, 1998.
95. Bowen J, Noakes M, Clifton PM. Appetite regulatory hormone responses to various
74. Berg C, Lappas G, Wolk A, Strandhagen E, Toren K, Rosengren A, Thelle D, Lissner dietary proteins differ by body mass index status despite similar reductions in ad
L. Eating patterns and portion size associated with obesity in a Swedish population. libitum energy intake. J Clin Endocrinol Metab 91: 2913–2919, 2006.
Appetite 52: 21–26, 2009.
96. Bowen J, Noakes M, Trenerry C, Clifton PM. Energy intake, ghrelin, and cholecysto-
75. Berthoud HR. Multiple neural systems controlling food intake and body weight. Neu- kinin after different carbohydrate and protein preloads in overweight men. J Clin
rosci Biobehav Rev 26: 393– 428, 2002. Endocrinol Metab 91: 1477–1483, 2006.
76. Berthoud HR. The neurobiology of food intake in an obesogenic environment. Proc 97. Boyd KA, O’Donovan DG, Doran S, Wishart J, Chapman IM, Horowitz M, Feinle C.
Nutr Soc 71: 478 – 487, 2012. High-fat diet effects on gut motility, hormone, and appetite responses to duodenal
lipid in healthy men. Am J Physiol Gastrointest Liver Physiol 284: G188 –G196, 2003.
77. Berthoud HR, Patterson LM. Anatomical relationship between vagal afferent fibers
and CCK-immunoreactive entero-endocrine cells in the rat small intestinal mucosa. 98. Breen DM, Rasmussen BA, Cote CD, Jackson VM, Lam TK. Nutrient-sensing mech-
Acta Anat 156: 123–131, 1996. anisms in the gut as therapeutic targets for diabetes. Diabetes 62: 3005–3013, 2013.
99. Breen DM, Yue JT, Rasmussen BA, Kokorovic A, Cheung GW, Lam TK. Duodenal 119. Burton-Freeman B, Davis PA, Schneeman BO. Interaction of fat availability and sex on
PKC-delta and cholecystokinin signaling axis regulates glucose production. Diabetes postprandial satiety and cholecystokinin after mixed-food meals. Am J Clin Nutr 80:
60: 3148 –3153, 2011. 1207–1214, 2004.
100. Brennan IM, Feltrin KL, Horowitz M, Smout AJ, Meyer JH, Wishart J, Feinle-Bisset C. 120. Cabrele C, Beck-Sickinger AG. Molecular characterization of the ligand-receptor
Evaluation of interactions between CCK and GLP-1 in their effects on appetite, interaction of the neuropeptide Y family. J Pept Sci 6: 97–122, 2000.
energy intake, and antropyloroduodenal motility in healthy men. Am J Physiol Regul
Integr Comp Physiol 288: R1477–R1485, 2005. 121. Calanna S, Christensen M, Holst JJ, Laferrere B, Gluud LL, Vilsboll T, Knop FK.
Secretion of glucagon-like peptide-1 in patients with type 2 diabetes mellitus: system-
101. Brennan IM, Feltrin KL, Nair NS, Hausken T, Little TJ, Gentilcore D, Wishart JM, Jones atic review and meta-analyses of clinical studies. Diabetologia 56: 965–972, 2013.
KL, Horowitz M, Feinle-Bisset C. Effects of the phases of the menstrual cycle on
gastric emptying, glycemia, plasma GLP-1 and insulin, and energy intake in healthy lean 122. Calbet JA, Holst JJ. Gastric emptying, gastric secretion and enterogastrone response
women. Am J Physiol Gastrointest Liver Physiol 297: G602–G610, 2009. after administration of milk proteins or their peptide hydrolysates in humans. Eur J
Nutr 43: 127–139, 2004.
102. Brennan IM, Little TJ, Feltrin KL, Smout AJ, Wishart JM, Horowitz M, Feinle-Bisset C.
Dose-dependent effects of cholecystokinin-8 on antropyloroduodenal motility, gas- 123. Calbet JA, MacLean DA. Role of caloric content on gastric emptying in humans. J
trointestinal hormones, appetite, and energy intake in healthy men. Am J Physiol Physiol 498: 553–559, 1997.
Endocrinol Metab 295: E1487–E1494, 2008.
124. Callahan HS, Cummings DE, Pepe MS, Breen PA, Matthys CC, Weigle DS. Postpran-
103. Brennan IM, Luscombe-Marsh ND, Seimon RV, Otto B, Horowitz M, Wishart JM, dial suppression of plasma ghrelin level is proportional to ingested caloric load but
Feinle-Bisset C. Effects of fat, protein and carbohydrate, and protein load, on appe- does not predict intermeal interval in humans. J Clin Endocrinol Metab 89: 1319 –1324,
tite, plasma cholecystokinin, peptide YY and ghrelin, and energy intake in lean and 2004.
obese men. Am J Physiol Gastrointest Liver Physiol 303: G129 –G140, 2012.
125. Cameron KO, Bhattacharya SK, Loomis AK. Small molecule ghrelin receptor inverse
104. Brennan IM, Otto B, Feltrin KL, Meyer JH, Horowitz M, Feinle-Bisset C. Intravenous
agonists and antagonists. J Med Chem 57: 8671– 8691, 2014.
CCK-8, but not GLP-1, suppresses ghrelin and stimulates PYY release in healthy men.
Peptides 28: 607– 611, 2007. 126. Camilleri M. Integrated upper gastrointestinal response to food intake. Gastroenter-
ology 131: 640 – 658, 2006.
105. Brighton CA, Rievaj J, Kuhre RE, Glass LL, Schoonjans K, Holst JJ, Gribble FM,
Reimann F. Bile acids trigger GLP-1 release predominantly by accessing basolaterally 127. Camilleri M, Parkman HP, Shafi MA, Abell TL, Gerson L, American College of Gas-
located G protein-coupled bile acid receptors. Endocrinology 156: 3961–3970, 2015. troenterology. Clinical guideline: management of gastroparesis. Am J Gastroenterol
108: 18 –37, 2013.
106. Broglio F, Arvat E, Benso A, Gottero C, Muccioli G, Papotti M, van der Lely AJ,
Deghenghi R, Ghigo E. Ghrelin, a natural GH secretagogue produced by the stomach, 128. Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin
induces hyperglycemia and reduces insulin secretion in humans. J Clin Endocrinol
hormone action. Cell Metab 17: 819 – 837, 2013.
Metab 86: 5083–5086, 2001.
129. Carbonnel F, Lemann M, Rambaud JC, Mundler O, Jian R. Effect of the energy density
107. Broglio F, Gottero C, Van Koetsveld P, Prodam F, Destefanis S, Benso A, Gauna C,
of a solid-liquid meal on gastric emptying and satiety. Am J Clin Nutr 60: 307–311,
Hofland L, Arvat E, van der Lely AJ, Ghigo E. Acetylcholine regulates ghrelin secretion
1994.
in humans. J Clin Endocrinol Metab 89: 2429 –2433, 2004.
130. Carr RD, Larsen MO, Jelic K, Lindgren O, Vikman J, Holst JJ, Deacon CF, Ahren B.
108. Brolin RE, Robertson LB, Kenler HA, Cody RP. Weight loss and dietary intake after
Secretion and dipeptidyl peptidase-4-mediated metabolism of incretin hormones af-
vertical banded gastroplasty and Roux-en-Y gastric bypass. Ann Surg 220: 782–790,
ter a mixed meal or glucose ingestion in obese compared with lean, nondiabetic men.
1994.
J Clin Endocrinol Metab 95: 872– 878, 2010.
109. Brown JC, Otte SC. Gastrointestinal hormones and the control of insulin secretion.
Diabetes 27: 782–787, 1978. 131. Carr RD, Larsen MO, Winzell MS, Jelic K, Lindgren O, Deacon CF, Ahren B. Incretin
and islet hormonal responses to fat and protein ingestion in healthy men. Am J Physiol
110. Brubaker PL, Anini Y. Direct and indirect mechanisms regulating secretion of gluca- Endocrinol Metab 295: E779 –E784, 2008.
gon-like peptide-1 and glucagon-like peptide-2. Can J Physiol Pharmacol 81: 1005–
1012, 2003. 132. Carroll JF, Kaiser KA, Franks SF, Deere C, Caffrey JL. Influence of BMI and gender on
postprandial hormone responses. Obesity 15: 2974 –2983, 2007.
111. Brunstrom JM. Mind over platter: pre-meal planning and the control of meal size in
humans. Int J Obes 38 Suppl 1: S9 –12, 2014. 133. Castiglione KE, Read NW, French SJ. Food intake responses to upper gastrointestinal
lipid infusions in humans. Physiol Behav 64: 141–145, 1998.
112. Bucceri AM, Calogero AE, Brogna A. Gallbladder and gastric emptying: relationship to
cholecystokininemia in diabetics. Eur J Intern Med 13: 123–128, 2002. 134. Cha R, Marescaux J, Diana M. Updates on gastric electrical stimulation to treat obe-
sity: systematic review and future perspectives. World J Gastrointest Endosc 6: 419 –
113. Bueter M, le Roux CW. Gastrointestinal hormones, energy balance and bariatric 431, 2014.
surgery. Int J Obes 35 Suppl 3: S35–39, 2011.
135. Chaiban JT, Bitar FF, Azar ST. Effect of chronic hypoxia on leptin, insulin, adiponectin,
114. Buffa R, Capella C, Fontana P, Usellini L, Solcia E. Types of endocrine cells in the and ghrelin. Metabolism 57: 1019 –1022, 2008.
human colon and rectum. Cell Tissue Res 192: 227–240, 1978.
136. Chaikomin R, Doran S, Jones KL, Feinle-Bisset C, O’Donovan D, Rayner CK, Horow-
115. Bullock BP, Heller RS, Habener JF. Tissue distribution of messenger ribonucleic acid
itz M. Initially more rapid small intestinal glucose delivery increases plasma insulin,
encoding the rat glucagon-like peptide-1 receptor. Endocrinology 137: 2968 –2978,
GIP, and GLP-1 but does not improve overall glycemia in healthy subjects. Am J Physiol
1996.
Endocrinol Metab 289: E504 –E507, 2005.
116. Burcelin R, Brunner H, Seydoux J, Thorensa B, Pedrazzini T. Increased insulin con-
137. Chaikomin R, Wu KL, Doran S, Jones KL, Smout AJ, Renooij W, Holloway RH, Meyer
centrations and glucose storage in neuropeptide Y Y1 receptor-deficient mice. Pep-
JH, Horowitz M, Rayner CK. Concurrent duodenal manometric and impedance re-
tides 22: 421– 427, 2001.
cording to evaluate the effects of hyoscine on motility and flow events, glucose ab-
117. Burcelin R, Da Costa A, Drucker D, Thorens B. Glucose competence of the hepato- sorption, and incretin release. Am J Physiol Gastrointest Liver Physiol 292: G1099 –
portal vein sensor requires the presence of an activated glucagon-like peptide-1 G1104, 2007.
receptor. Diabetes 50: 1720 –1728, 2001.
138. Chambers AP, Jessen L, Ryan KK, Sisley S, Wilson-Perez HE, Stefater MA, Gaitonde
118. Burton DD, Kim HJ, Camilleri M, Stephens DA, Mullan BP, O’Connor MK, Talley NJ. SG, Sorrell JE, Toure M, Berger J, D’Alessio DA, Woods SC, Seeley RJ, Sandoval DA.
Relationship of gastric emptying and volume changes after a solid meal in humans. Am Weight-independent changes in blood glucose homeostasis after gastric bypass or
J Physiol Gastrointest Liver Physiol 289: G261–G266, 2005. vertical sleeve gastrectomy in rats. Gastroenterology 141: 950 –958, 2011.
139. Chan JL, Bullen J, Lee JH, Yiannakouris N, Mantzoros CS. Ghrelin levels are not 159. Colombel JF, Sutton A, Chayvialle JA, Modigliani R. Cholecystokinin release and bil-
regulated by recombinant leptin administration and/or three days of fasting in healthy iopancreatic secretion in response to selective perfusion of the duodenal loop with
subjects. J Clin Endocrinol Metab 89: 335–343, 2004. aminoacids in man. Gut 29: 1158 –1166, 1988.
140. Chandarana K, Gelegen C, Irvine EE, Choudhury AI, Amouyal C, Andreelli F, Withers 160. Conigrave AD, Hampson DR. Broad-spectrum amino acid-sensing class C G-protein
DJ, Batterham RL. Peripheral activation of the Y2-receptor promotes secretion of coupled receptors: molecular mechanisms, physiological significance and options for
GLP-1 and improves glucose tolerance. Mol Metab 2: 142–152, 2013. drug development. Pharmacol Ther 127: 252–260, 2010.
141. Chandarana K, Gelegen C, Karra E, Choudhury AI, Drew ME, Fauveau V, Viollet B, 161. Conigrave AD, Quinn SJ, Brown EM. L-Amino acid sensing by the extracellular Ca2⫹-
Andreelli F, Withers DJ, Batterham RL. Diet and gastrointestinal bypass-induced sensing receptor. Proc Natl Acad Sci USA 97: 4814 – 4819, 2000.
weight loss: the roles of ghrelin and peptide YY. Diabetes 60: 810 – 818, 2011.
162. Corvilain B, Abramowicz M, Fery F, Schoutens A, Verlinden M, Balasse E, Horowitz
142. Chang J, Wu T, Greenfield JR, Samocha-Bonet D, Horowitz M, Rayner CK. Effects of M. Effect of short-term starvation on gastric emptying in humans: relationship to oral
intraduodenal glutamine on incretin hormone and insulin release, the glycemic re- glucose tolerance. Am J Physiol Gastrointest Liver Physiol 269: G512–G517, 1995.
sponse to an intraduodenal glucose infusion, and antropyloroduodenal motility in
163. Cote CD, Zadeh-Tahmasebi M, Rasmussen BA, Duca FA, Lam TK. Hormonal signal-
health and type 2 diabetes. Diabetes Care 36: 2262–2265, 2013.
ing in the gut. J Biol Chem 289: 11642–11649, 2014.
143. Chelikani PK, Haver AC, Reidelberger RD. Dose-dependent effects of peptide YY(3– 164. Cowley MA, Smith RG, Diano S, Tschop M, Pronchuk N, Grove KL, Strasburger CJ,
36) on conditioned taste aversion in rats. Peptides 27: 3193–3201, 2006. Bidlingmaier M, Esterman M, Heiman ML, Garcia-Segura LM, Nillni EA, Mendez P,
Low MJ, Sotonyi P, Friedman JM, Liu H, Pinto S, Colmers WF, Cone RD, Horvath TL.
144. Chelikani PK, Haver AC, Reidelberger RD. Intravenous infusion of peptide YY(3–36)
The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel
potently inhibits food intake in rats. Endocrinology 146: 879 – 888, 2005.
hypothalamic circuit regulating energy homeostasis. Neuron 37: 649 – 661, 2003.
145. Chen HY, Trumbauer ME, Chen AS, Weingarth DT, Adams JR, Frazier EG, Shen Z,
165. Cox JE. Cholecystokinin satiety involves CCKA receptors perfused by the superior
Marsh DJ, Feighner SD, Guan XM, Ye Z, Nargund RP, Smith RG, Van der Ploeg LH,
pancreaticoduodenal artery. Am J Physiol Regul Integr Comp Physiol 274: R1390 –
Howard AD, MacNeil DJ, Qian S. Orexigenic action of peripheral ghrelin is mediated
R1396, 1998.
by neuropeptide Y and agouti-related protein. Endocrinology 145: 2607–2612, 2004.
166. Creutzfeldt W, Gregory RA, Grossman MI, Pearse AGE. Origin, Chemistry, Physiol-
146. Chen JH, Song GQ, Yin J, Sun Y, Chen JD. Gastric electrical stimulation reduces ogy and Pathophysiology of the Gastrointestinal Hormones. Proc Int Symp Wiesbaden
visceral sensitivity to gastric distention in healthy canines. Auton Neurosci 160: 16 –20, 1969: 95, 1970.
2011.
167. Crum AJ, Corbin WR, Brownell KD, Salovey P. Mind over milkshakes: mindsets, not
147. Cheung GWC, Kokorovic A, Lam CKL, Chari M, Lam TKT. Intestinal cholecystokinin just nutrients, determine ghrelin response. Health Psychol 30: 424 – 429, 2011.
controls glucose production through a neuronal network. Cell Metab 10: 99 –109,
2009. 168. Cuber JC, Bernard G, Fushiki T, Bernard C, Yamanishi R, Sugimoto E, Chayvialle JA.
Luminal CCK-releasing factors in the isolated vascularly perfused rat duodenojeju-
148. Cho HJ, Kosari S, Hunne B, Callaghan B, Rivera LR, Bravo DM, Furness JB. Differences num. Am J Physiol Gastrointest Liver Physiol 259: G191–G197, 1990.
in hormone localisation patterns of K and L type enteroendocrine cells in the mouse
and pig small intestine and colon. Cell Tissue Res 359: 693– 698, 2015. 169. Cukier K, Pilichiewicz AN, Chaikomin R, Brennan IM, Wishart JM, Rayner CK, Jones
KL, Horowitz M, Feinle-Bisset C. Effect of small intestinal glucose load on plasma
149. Choi S, Lee M, Shiu AL, Yo SJ, Halldén G, Aponte GW. GPR93 activation by protein ghrelin in healthy men. Am J Physiol Regul Integr Comp Physiol 295: R459 –R462, 2008.
hydrolysate induces CCK transcription and secretion in STC-1 cells. Am J Physiol
Gastrointest Liver Physiol 292: G1366 –G1375, 2007. 170. Cummings DE, Foster-Schubert KE, Overduin J. Ghrelin and energy balance: focus on
current controversies. Curr Drug Targets 6: 153–169, 2005.
150. Christensen M, Sparre-Ulrich AH, Hartmann B, Grevstad U, Rosenkilde MM, Holst JJ,
Vilsboll T, Knop FK. Transfer of liraglutide from blood to cerebrospinal fluid is minimal 171. Cummings DE, Frayo RS, Marmonier C, Aubert R, Chapelot D. Plasma ghrelin levels
in patients with type 2 diabetes. Int J Obes 39: 1651–1654, 2015. and hunger scores in humans initiating meals voluntarily without time- and food-
related cues. Am J Physiol Endocrinol Metab 287: E297–E304, 2004.
151. Chu ZL, Carroll C, Alfonso J, Gutierrez V, He H, Lucman A, Pedraza M, Mondala H,
172. Cummings DE, Overduin J. Gastrointestinal regulation of food intake. J Clin Invest 117:
Gao H, Bagnol D, Chen R, Jones RM, Behan DP, Leonard J. A role for intestinal
13–23, 2007.
endocrine cell-expressed G protein-coupled receptor 119 in glycemic control by
enhancing glucagon-like peptide-1 and glucose-dependent insulinotropic peptide re- 173. Cummings DE, Purnell JQ, Frayo RS, Schmidova K, Wisse BE, Weigle DS. A prepran-
lease. Endocrinology 149: 2038 –2047, 2008. dial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes
50: 1714 –1719, 2001.
152. Chuang JC, Sakata I, Kohno D, Perello M, Osborne-Lawrence S, Repa JJ, Zigman JM.
Ghrelin directly stimulates glucagon secretion from pancreatic alpha-cells. Mol Endo- 174. Cummings DE, Weigle DS, Frayo RS, Breen PA, Ma MK, Dellinger EP, Purnell JQ.
crinol 25: 1600 –1611, 2011. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J
Med 346: 1623–1630, 2002.
153. Cigaina V, Hirschberg AL. Gastric pacing for morbid obesity: plasma levels of gastro-
intestinal peptides and leptin. Obes Res 11: 1456 –1462, 2003. 175. Cunningham KM, Daly J, Horowitz M, Read NW. Gastrointestinal adaptation to diets
of differing fat composition in human volunteers. Gut 32: 483– 486, 1991.
154. Cigaina V, Hirschberg AL. Plasma ghrelin and gastric pacing in morbidly obese pa-
tients. Metabolism 56: 1017–1021, 2007. 176. Cunningham KM, Horowitz M, Read NW. The effect of short-term dietary supple-
mentation with glucose on gastric emptying in humans. Br J Nutr 65: 15–19, 1991.
155. Cohen DA. Neurophysiological pathways to obesity: below awareness and beyond
individual control. Diabetes 57: 1768 –1773, 2008. 177. Cushing CC, Benoit SC, Peugh JL, Reiter-Purtill J, Inge TH, Zeller MH. Longitudinal
trends in hedonic hunger after Roux-en-Y gastric bypass in adolescents. Surg Obes
156. Cohen S. “What is physiological?” An Answer! Gastroenterology 66 4791768 – 480, Relat Dis 10: 125–130, 2014.
1974.
178. D’Alessio D, Lu W, Sun W, Zheng S, Yang Q, Seeley R, Woods SC, Tso P. Fasting and
157. Cole AJ, Teigen LM, Jahansouz C, Earthman CP, Sibley SD. The influence of bariatric postprandial concentrations of GLP-1 in intestinal lymph and portal plasma: evidence
surgery on serum bile acids in humans and potential metabolic and hormonal impli- for selective release of GLP-1 in the lymph system. Am J Physiol Regul Integr Comp
cations: a systematic review. Curr Obes Rep 4: 441– 450, 2015. Physiol 293: R2163–R2169, 2007.
158. Collins PJ, Horowitz M, Cook DJ, Harding PE, Shearman DJ. Gastric emptying in 179. Dailey MJ, Moghadam AA, Moran TH. Jejunal linoleic acid infusions require GLP-1
normal subjects–a reproducible technique using a single scintillation camera and com- receptor signaling to inhibit food intake: implications for the effectiveness of
puter system. Gut 24: 1117–1125, 1983. Roux-en-Y gastric bypass. Am J Physiol Endocrinol Metab 301: E1184 –E1190, 2011.
180. Dailey MJ, Stingl KC, Moran TH. Disassociation between preprandial gut peptide 200. Degen LP, Phillips SF. Variability of gastrointestinal transit in healthy women and men.
release and food-anticipatory activity. Endocrinology 153: 132–142, 2012. Gut 39: 299 –305, 1996.
181. Daly DM, Park SJ, Valinsky WC, Beyak MJ. Impaired intestinal afferent nerve satiety 201. Delgado-Aros S, Camilleri M, Castillo EJ, Cremonini F, Stephens D, Ferber I, Baxter K,
signalling and vagal afferent excitability in diet induced obesity in the mouse. J Physiol Burton D, Zinsmeister AR. Effect of gastric volume or emptying on meal-related
589: 2857–2870, 2011. symptoms after liquid nutrients in obesity: a pharmacologic study. Clin Gastroenterol
Hepatol 3: 997–1006, 2005.
182. Daly K, Al-Rammahi M, Moran A, Marcello M, Ninomiya Y, Shirazi-Beechey SP.
Sensing of amino acids by the gut-expressed taste receptor T1R1-T1R3 stimulates 202. Delhanty PJ, van der Lely AJ. Ghrelin and glucose homeostasis. Peptides 32: 2309 –
CCK secretion. Am J Physiol Gastrointest Liver Physiol 304: G271–G282, 2013. 2318, 2011.
183. Darcel NP, Liou AP, Tome D, Raybould HE. Activation of vagal afferents in the rat 203. Deloose E, Janssen P, Depoortere I, Tack J. The migrating motor complex: control
duodenum by protein digests requires PepT1. J Nutr 135: 1491–1495, 2005. mechanisms and its role in health and disease. Nat Rev Gastroenterol Hepatol 9: 271–
285, 2012.
184. Date Y, Kojima M, Hosoda H, Sawaguchi A, Mondal MS, Suganuma T, Matsukura S,
Kangawa K, Nakazato M. Ghrelin, a novel growth hormone-releasing acylated pep- 204. Deloose E, Vos R, Corsetti M, Depoortere I, Tack J. Endogenous motilin, but not
tide, is synthesized in a distinct endocrine cell type in the gastrointestinal tracts of rats ghrelin plasma levels fluctuate in accordance with gastric phase III activity of the
and humans. Endocrinology 141: 4255– 4261, 2000. migrating motor complex in man. Neurogastroenterol Motil 27: 63–71, 2015.
185. Date Y, Murakami N, Toshinai K, Matsukura S, Niijima A, Matsuo H, Kangawa K, 205. Deloose E, Vos R, Janssen P, Van den Bergh O, Van Oudenhove L, Depoortere I, Tack
Nakazato M. The role of the gastric afferent vagal nerve in ghrelin-induced feeding and J. The motilin receptor agonist erythromycin stimulates hunger and food intake
growth hormone secretion in rats. Gastroenterology 123: 1120 –1128, 2002. through a cholinergic pathway. Am J Clin Nutr 103: 730 –737, 2016.
186. Date Y, Toshinai K, Koda S, Miyazato M, Shimbara T, Tsuruta T, Niijima A, Kangawa 206. Delzenne N, Blundell J, Brouns F, Cunningham K, De Graaf K, Erkner A, Lluch A, Mars
K, Nakazato M. Peripheral interaction of ghrelin with cholecystokinin on feeding M, Peters HP, Westerterp-Plantenga M. Gastrointestinal targets of appetite regula-
regulation. Endocrinology 146: 3518 –3525, 2005. tion in humans. Obes Rev 11: 234 –250, 2010.
187. Datz FL, Christian PE, Moore J. Gender-related differences in gastric emptying. J Nucl 207. Depoortere I. Taste receptors of the gut: emerging roles in health and disease. Gut 63:
Med 28: 1204 –1207, 1987. 179 –190, 2014.
188. Davis JD, Smith GP. Learning to sham feed: behavioral adjustments to loss of physio- 208. Dezaki K, Damdindorj B, Sone H, Dyachok O, Tengholm A, Gylfe E, Kurashina T,
logical postingestional stimuli. Am J Physiol Regul Integr Comp Physiol 259: R1228 – Yoshida M, Kakei M, Yada T. Ghrelin attenuates cAMP-PKA signaling to evoke insu-
R1235, 1990. linostatic cascade in islet beta-cells. Diabetes 60: 2315–2324, 2011.
189. De Castro JM, King GA, Duarte-Gardea M, Gonzalez-Ayala S, Kooshian CH. Over- 209. Dhillo WS, Murphy KG, Bloom S. Endocrinology: the next 60 years. J Endocrinol 190:
weight and obese humans overeat away from home. Appetite 59: 204 –211, 2012. 7–10, 2006.
190. De Giorgio R, Stanghellini V, Ricci Maccarini M, Morselli-Labate AM, Barbara G, 210. Diakogiannaki E, Pais R, Tolhurst G, Parker HE, Horscroft J, Rauscher B, Zietek T,
Franzoso L, Rovati LC, Corinaldesi R, Barbara L, Go VL. Effects of dietary fat on Daniel H, Gribble FM, Reimann F. Oligopeptides stimulate glucagon-like peptide-1
postprandial gastrointestinal motility are inhibited by a cholecystokinin type A recep- secretion in mice through proton-coupled uptake and the calcium-sensing receptor.
tor antagonist. Ann NY Acad Sci 713: 226 –231, 1994. Diabetologia 56: 2688 –2696, 2013.
191. De Hollanda A, Casals G, Delgado S, Jimenez A, Viaplana J, Lacy AM, Vidal J. Gastro- 211. Dickson SL, Egecioglu E, Landgren S, Skibicka KP, Engel JA, Jerlhag E. The role of the
intestinal hormones and weight loss maintenance following Roux-en-Y gastric bypass. central ghrelin system in reward from food and chemical drugs. Mol Cell Endocrinol
J Clin Endocrinol Metab 100: 4677– 4684, 2015. 340: 80 – 87, 2011.
192. De Krom M, van der Schouw YT, Hendriks J, Ophoff RA, van Gils CH, Stolk RP, 212. Dickson SL, Shirazi RH, Hansson C, Bergquist F, Nissbrandt H, Skibicka KP. The
Grobbee DE, Adan R. Common genetic variations in CCK, leptin, and leptin receptor glucagon-like peptide 1 (GLP-1) analogue, exendin-4, decreases the rewarding value
genes are associated with specific human eating patterns. Diabetes 56: 276 –280, of food: a new role for mesolimbic GLP-1 receptors. J Neurosci 32: 4812– 4820, 2012.
2007.
213. Dirksen C, Damgaard M, Bojsen-Moller KN, Jorgensen NB, Kielgast U, Jacobsen SH,
193. De Lartigue G, Barbier de la Serre C, Espero E, Lee J, Raybould HE. Leptin resistance Naver LS, Worm D, Holst JJ, Madsbad S, Hansen DL, Madsen JL. Fast pouch emptying,
in vagal afferent neurons inhibits cholecystokinin signaling and satiation in diet induced delayed small intestinal transit, and exaggerated gut hormone responses after Roux-
obese rats. PLoS One 7: e32967 2012. en-Y gastric bypass. Neurogastroenterol Motil 25: 346 – e255, 2013.
194. De Silva A, Salem V, Long CJ, Makwana A, Newbould RD, Rabiner EA, Ghatei MA, 214. Dirksen C, Jorgensen NB, Bojsen-Moller KN, Kielgast U, Jacobsen SH, Clausen TR,
Bloom SR, Matthews PM, Beaver JD, Dhillo WS. The gut hormones PYY 3–36 and Worm D, Hartmann B, Rehfeld JF, Damgaard M, Madsen JL, Madsbad S, Holst JJ,
GLP-1 7–36 amide reduce food intake and modulate brain activity in appetite centers Hansen DL. Gut hormones, early dumping and resting energy expenditure in patients
in humans. Cell Metab 14: 700 –706, 2011. with good and poor weight loss response after Roux-en-Y gastric bypass. Int J Obes 37:
1452–1459, 2013.
195. Deacon CF, Ahren B. Physiology of incretins in health and disease. Rev Diabet Stud 8:
293–306, 2011. 215. Distrutti E, Azpiroz F, Soldevilla A, Malagelada JR. Gastric wall tension determines
perception of gastric distention. Gastroenterology 116: 1035–1042, 1999.
196. Deane AM, Nguyen NQ, Stevens JE, Fraser RJ, Holloway RH, Besanko LK, Burgstad
C, Jones KL, Chapman MJ, Rayner CK, Horowitz M. Endogenous glucagon-like pep- 216. Dockray GJ. Cholecystokinin. Curr Opin Endocrinol Diabetes Obes 19: 8 –12, 2012.
tide-1 slows gastric emptying in healthy subjects, attenuating postprandial glycemia. J
Clin Endocrinol Metab 95: 215–221, 2010. 217. Dockray GJ. Gastrointestinal hormones and the dialogue between gut and brain. J
Physiol 592: 2927–2941, 2014.
197. Degen L, Drewe J, Piccoli F, Grani K, Oesch S, Bunea R, D’Amato M, Beglinger C.
Effect of CCK-1 receptor blockade on ghrelin and PYY secretion in men. Am J Physiol 218. Doisy EA. Sex Hormones. The Porter Lectures, 1936. Lawrence, KS: Univ. of Kansas
Regul Integr Comp Physiol 292: R1391–R1399, 2007. Press, 1936.
198. Degen L, Oesch S, Casanova M, Graf S, Ketterer S, Drewe J, Beglinger C. Effect of 219. Dossat AM, Diaz R, Gallo L, Panagos A, Kay K, Williams DL. Nucleus accumbens
peptide YY3-36 on food intake in humans. Gastroenterology 129: 1430 –1436, 2005. GLP-1 receptors influence meal size and palatability. Am J Physiol Endocrinol Metab
304: E1314 –E1320, 2013.
199. Degen L, Oesch S, Matzinger D, Drewe J, Knupp M, Zimmerli F, Beglinger C. Effects
of a preload on reduction of food intake by GLP-1 in healthy subjects. Digestion 74: 220. Drozdzal M, Segui S, Radeva P, Malagelada C, Azpiroz F, Vitria J. Motility bar: a new
78 – 84, 2006. tool for motility analysis of endoluminal videos. Comput Biol Med 65: 320 –330, 2015.
221. Druce MR, Wren AM, Park AJ, Milton JE, Patterson M, Frost G, Ghatei MA, Small C, 240. Erdmann J, Lippl F, Wagenpfeil S, Schusdziarra V. Differential association of basal and
Bloom SR. Ghrelin increases food intake in obese as well as lean subjects. Int J Obes 29: postprandial plasma ghrelin with leptin, insulin, and type 2 diabetes. Diabetes 54:
1130 –1136, 2005. 1371–1378, 2005.
222. Drucker DJ. The biology of incretin hormones. Cell Metab 3: 153–165, 2006. 241. Essah PA, Levy JR, Sistrun SN, Kelly SM, Nestler JE. Effect of macronutrient compo-
sition on postprandial peptide YY levels. J Clin Endocrinol Metab 92: 4052– 4055, 2007.
223. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor ago-
nists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368: 1696 –1705, 242. Eysselein VE, Eberlein GA, Hesse WH, Schaeffer M, Grandt D, Williams R, Goebell H,
2006. Reeve JR Jr. Molecular variants of cholecystokinin after endogenous stimulation in
humans: a time study. Am J Physiol Gastrointest Liver Physiol 258: G951–G957, 1990.
224. Duca FA, Sakar Y, Covasa M. Combination of obesity and high-fat feeding diminishes
sensitivity to GLP-1R agonist exendin-4. Diabetes 62: 2410 –2415, 2013. 243. Eysselein VE, Eberlein GA, Schaeffer M, Grandt D, Goebell H, Niebel W, Rosenquist
GL, Meyer HE, Reeve JR Jr. Characterization of the major form of cholecystokinin in
225. Duca FA, Zhong L, Covasa M. Reduced CCK signaling in obese-prone rats fed a high human intestine: CCK-58. Am J Physiol Gastrointest Liver Physiol 258: G253–G260,
fat diet. Horm Behav 64: 812– 817, 2013. 1990.
226. Dumont Y, Fournier A, St-Pierre S, Quirion R. Characterization of neuropeptide Y 244. Falken Y, Hellstrom PM, Holst JJ, Naslund E. Changes in glucose homeostasis after
binding sites in rat brain membrane preparations using [125I][Leu31,Pro34]peptide YY Roux-en-Y gastric bypass surgery for obesity at day three, two months, and one year
and [125I]peptide YY3-36 as selective Y1 and Y2 radioligands. J Pharmacol Exp Ther after surgery: role of gut peptides. J Clin Endocrinol Metab 96: 2227–2235, 2011.
272: 673– 680, 1995.
245. Feinle C, Chapman IM, Wishart J, Horowitz M. Plasma glucagon-like peptide-1
227. Dumoulin V, Dakka T, Plaisancie P, Chayvialle JA, Cuber JC. Regulation of glucagon- (GLP-1) responses to duodenal fat and glucose infusions in lean and obese men.
like peptide-1-(7–36) amide, peptide YY, and neurotensin secretion by neurotrans- Peptides 23: 1491–1495, 2002.
mitters and gut hormones in the isolated vascularly perfused rat ileum. Endocrinology
136: 5182–5188, 1995. 246. Feinle C, D’Amato M, Read NW. Cholecystokinin-A receptors modulate gastric sen-
sory and motor responses to gastric distension and duodenal lipid. Gastroenterology
228. Dyer J, Salmon KSH, Zibrik L, Shirazi-Beechey SP. Expression of sweet taste receptors 110: 1379 –1385, 1996.
of the T1R family in the intestinal tract and enteroendocrine cells. Biochem Soc Trans 33:
302–305, 2005. 247. Feinle C, O’Donovan D, Doran S, Andrews JM, Wishart J, Chapman I, Horowitz M.
Effects of fat digestion on appetite, APD motility, and gut hormones in response to
229. Edkins JS. The chemical mechanism of gastric secretion. J Physiol 34: 133–144, 1906. duodenal fat infusion in humans. Am J Physiol Gastrointest Liver Physiol 284: G798 –
G807, 2003.
230. Edwards CM, Todd JF, Mahmoudi M, Wang Z, Wang RM, Ghatei MA, Bloom SR.
Glucagon-like peptide 1 has a physiological role in the control of postprandial glucose 248. Feinle-Bisset C, Patterson M, Ghatei MA, Bloom SR, Horowitz M. Fat digestion is
in humans: studies with the antagonist exendin 9 –39. Diabetes 48: 86 –93, 1999. required for suppression of ghrelin and stimulation of peptide YY and pancreatic
polypeptide secretion by intraduodenal lipid. Am J Physiol Endocrinol Metab 289:
231. Egerod KL, Engelstoft MS, Grunddal KV, Nohr MK, Secher A, Sakata I, Pedersen J, E948 –E953, 2005.
Windelov JA, Fuchtbauer EM, Olsen J, Sundler F, Christensen JP, Wierup N, Olsen JV,
Holst JJ, Zigman JM, Poulsen SS, Schwartz TW. A major lineage of enteroendocrine 249. Feltrin KL, Little TJ, Meyer JH, Horowitz M, Smout AJPM, Wishart J, Pilichiewicz AN,
cells coexpress CCK, secretin, GIP, GLP-1, PYY, and neurotensin but not somatosta- Rades T, Chapman IM, Feinle-Bisset C. Effects of intraduodenal fatty acids on appe-
tin. Endocrinology 153: 5782–5795, 2012. tite, antropyloroduodenal motility, and plasma CCK and GLP-1 in humans vary with
their chain length. Am J Physiol Regul Integr Comp Physiol 287: R524 –R533, 2004.
232. Eisen S, Phillips RJ, Geary N, Baronowsky EA, Powley TL, Smith GP. Inhibitory effects
on intake of cholecystokinin-8 and cholecystokinin-33 in rats with hepatic proper or 250. Feltrin KL, Patterson M, Ghatei MA, Bloom SR, Meyer JH, Horowitz M, Feinle-Bisset
common hepatic branch vagal innervation. Am J Physiol Regul Integr Comp Physiol 289: C. Effect of fatty acid chain length on suppression of ghrelin and stimulation of PYY,
R456 –R462, 2005. GLP-2 and PP secretion in healthy men. Peptides 27: 1638 –1643, 2006.
233. Eissele R, Goke R, Willemer S, Harthus HP, Vermeer H, Arnold R, Goke B. Glucagon- 251. Fenske WK, Bueter M, Miras AD, Ghatei MA, Bloom SR, le Roux CW. Exogenous
like peptide-1 cells in the gastrointestinal tract and pancreas of rat, pig and man. Eur J peptide YY3-36 and Exendin-4 further decrease food intake, whereas octreotide
increases food intake in rats after Roux-en-Y gastric bypass. Int J Obes 36: 379 –384,
Clin Invest 22: 283–291, 1992.
2012.
234. Elashoff JD, Reedy TJ, Meyer JH. Analysis of gastric emptying data. Gastroenterology
252. Fernandez-Garcia JC, Murri M, Coin-Araguez L, Alcaide J, El Bekay R, Tinahones FJ.
83: 1306 –1312, 1982.
GLP-1 and peptide YY secretory response after fat load is impaired by insulin resis-
235. Elliott RM, Morgan LM, Tredger JA, Deacon S, Wright J, Marks V. Glucagon-like tance, impaired fasting glucose and type 2 diabetes in morbidly obese subjects. Clin
peptide-1 (7–36)amide and glucose-dependent insulinotropic polypeptide secretion Endocrinol 80: 671– 676, 2014.
in response to nutrient ingestion in man: acute post-prandial and 24-h secretion
253. Fieseler P, Bridenbaugh S, Nustede R, Martell J, Orskov C, Holst JJ, Nauck MA.
patterns. J Endocrinol 138: 159 –166, 1993.
Physiological augmentation of amino acid-induced insulin secretion by GIP and GLP-I
236. Enc FY, Imeryuz N, Akin L, Turoglu T, Dede F, Haklar G, Tekesin N, Bekiroglu N, but not by CCK-8. Am J Physiol Endocrinol Metab 268: E949 –E955, 1995.
Yegen BC, Rehfeld JF, Holst JJ, Ulusoy NB. Inhibition of gastric emptying by acarbose
254. Flanagan DE, Evans ML, Monsod TP, Rife F, Heptulla RA, Tamborlane WV, Sherwin
is correlated with GLP-1 response and accompanied by CCK release. Am J Physiol
RS. The influence of insulin on circulating ghrelin. Am J Physiol Endocrinol Metab 284:
Gastrointest Liver Physiol 281: G752–G763, 2001.
E313–E316, 2003.
237. Engelstoft MS, Park WM, Sakata I, Kristensen LV, Husted AS, Osborne-Lawrence S,
255. Flint A, Raben A, Astrup A, Holst JJ. Glucagon-like peptide 1 promotes satiety and
Piper PK, Walker AK, Pedersen MH, Nohr MK, Pan J, Sinz CJ, Carrington PE, Akiyama
suppresses energy intake in humans. J Clin Invest 101: 515–520, 1998.
TE, Jones RM, Tang C, Ahmed K, Offermanns S, Egerod KL, Zigman JM, Schwartz
TW. Seven transmembrane G protein-coupled receptor repertoire of gastric ghrelin 256. Flint A, Raben A, Ersboll AK, Holst JJ, Astrup A. The effect of physiological levels of
cells. Mol Metab 2: 376 –392, 2013. glucagon-like peptide-1 on appetite, gastric emptying, energy and substrate metabo-
lism in obesity. Int J Obes Relat Metab Disord 25: 781–792, 2001.
238. English PJ, Ashcroft A, Patterson M, Dovey TM, Halford JC, Harrison J, Eccleston D,
Bloom SR, Ghatei MA, Wilding JP. Fasting plasma peptide-YY concentrations are 257. Flynn CR, Albaugh VL, Cai S, Cheung-Flynn J, Williams PE, Brucker RM, Bordenstein
elevated but do not rise postprandially in type 2 diabetes. Diabetologia 49: 2219 – SR, Guo Y, Wasserman DH, Abumrad NN. Bile diversion to the distal small intestine
2221, 2006. has comparable metabolic benefits to bariatric surgery. Nat Commun 6: 7715, 2015.
239. English PJ, Ghatei MA, Malik IA, Bloom SR, Wilding JP. Food fails to suppress ghrelin 258. Foster-Schubert KE, Overduin J, Prudom CE, Liu J, Callahan HS, Gaylinn BD, Thorner
levels in obese humans. J Clin Endocrinol Metab 87: 2984, 2002. MO, Cummings DE. Acyl and total ghrelin are suppressed strongly by ingested pro-
teins, weakly by lipids, and biphasically by carbohydrates. J Clin Endocrinol Metab 93: 278. Gibbs J, Young RC, Smith GP. Cholecystokinin decreases food intake in rats. J Comp
1971–1979, 2008. Physiol Psychol 84: 488 – 495, 1973.
259. Fraser R, Fone D, Horowitz M, Dent J. Cholecystokinin octapeptide stimulates phasic 279. Glatzle J, Raybould HE, Kueper MA, Reeve JR Jr, Zittel TT. Cholecystokinin-58 is
and tonic pyloric motility in healthy humans. Gut 34: 33–37, 1993. more potent in inhibiting food intake than cholecystokinin-8 in rats. Nutr Neurosci 11:
69 –74, 2008.
260. French SJ, Murray B, Rumsey RD, Sepple CP, Read NW. Is cholecystokinin a satiety
hormone? Correlations of plasma cholecystokinin with hunger, satiety and gastric 280. Glavas MM, Grayson BE, Allen SE, Copp DR, Smith MS, Cowley MA, Grove KL.
emptying in normal volunteers. Appetite 21: 95–104, 1993. Characterization of brainstem peptide YY (PYY) neurons. J Comp Neurol 506: 194 –
210, 2008.
261. French SJ, Murray B, Rumsey RD, Sepple CP, Read NW. Preliminary studies on the
gastrointestinal responses to fatty meals in obese people. Int J Obes Relat Metab Disord 281. Goebel-Stengel M, Stengel A, Wang L, Ohning G, Tache Y, Reeve JR Jr. CCK-8 and
17: 295–300, 1993. CCK-58 differ in their effects on nocturnal solid meal pattern in undisturbed rats. Am
J Physiol Regul Integr Comp Physiol 303: R850 –R860, 2012.
262. Fried M, Erlacher U, Schwizer W, Lochner C, Koerfer J, Beglinger C, Jansenr JB,
Lamers CB, Harder F, Bischof-Delaloye A, et al. Role of cholecystokinin in the regu- 282. Goetze O, Steingoetter A, Menne D, van der Voort IR, Kwiatek Ma Boesiger P,
lation of gastric emptying and pancreatic enzyme secretion in humans Studies with the Weishaupt D, Thumshirn M, Fried M, Schwizer W. The effect of macronutrients on
cholecystokinin-receptor antagonist loxiglumide. Gastroenterology 101: 503–511, gastric volume responses and gastric emptying in humans: a magnetic resonance
1991. imaging study. Am J Physiol Gastrointest Liver Physiol 292: G11–G17, 2007.
263. Fried M, Schwizer W, Beglinger C, Keller U, Jansen JB, Lamers CB. Physiological role 283. Goldstone AP, Miras AD, Scholtz S, Jackson S, Neff KJ, Penicaud L, Geoghegan J,
of cholecystokinin on postprandial insulin secretion and gastric meal emptying in man. Chhina N, Durighel G, Bell JD, Meillon S, le Roux CW. Link between increased satiety
Studies with the cholecystokinin receptor antagonist loxiglumide. Diabetologia 34: gut hormones and reduced food reward after gastric bypass surgery for obesity. J Clin
721–726, 1991. Endocrinol Metab 101: 599 – 609, 2016.
264. Fu-Cheng X, Anini Y, Chariot J, Castex N, Galmiche JP, Roze C. Mechanisms of
284. Goldstone AP, Prechtl CG, Scholtz S, Miras AD, Chhina N, Durighel G, Deliran SS,
peptide YY release induced by an intraduodenal meal in rats: neural regulation by
Beckmann C, Ghatei MA, Ashby DR, Waldman AD, Gaylinn BD, Thorner MO, Frost
proximal gut. Pflügers Arch 433: 571–579, 1997.
GS, Bloom SR, Bell JD. Ghrelin mimics fasting to enhance human hedonic, orbitofron-
265. Fuessl HS, Adrian TE, Uttenthal LO, Bloom SR. Peptide YY in diabetics treated tal cortex, and hippocampal responses to food. Am J Clin Nutr 99: 1319 –1330, 2014.
chronically with an intestinal glucosidase inhibitor. Klin Wochenschr 66: 985–989,
285. Gonlachanvit S, Hsu CW, Boden GH, Knight LC, Maurer AH, Fisher RS, Parkman HP.
1988.
Effect of altering gastric emptying on postprandial plasma glucose concentrations
266. Fujino K, Inui A, Asakawa A, Kihara N, Fujimura M, Fujimiya M. Ghrelin induces fasted following a physiologic meal in type-II diabetic patients. Dig Dis Sci 48: 488 – 497, 2003.
motor activity of the gastrointestinal tract in conscious fed rats. J Physiol 550: 227–240,
286. Goodwin ML, Harris JE, Hernandez A, Gladden LB. Blood lactate measurements and
2003.
analysis during exercise: a guide for clinicians. J Diabetes Sci Technol 1: 558 –569, 2007.
267. Geary N. Endocrine controls of eating: CCK, leptin, and ghrelin. Physiol Behav 81:
287. Gores GJ, Kost LJ, Miller LJ, LaRusso NF. Processing of cholecystokinin by isolated
719 –733, 2004.
liver cells. Am J Physiol Gastrointest Liver Physiol 257: G242–G248, 1989.
268. Geary N. Understanding synergy. Am J Physiol Endocrinol Metab 304: E237–E253,
2013. 288. Graham L, Murty G, Bowrey DJ. Taste, smell and appetite change after Roux-en-Y
gastric bypass surgery. Obes Surg 24: 1463–1468, 2014.
269. Geary N, Kissileff HR, Pi-Sunyer FX, Hinton V. Individual, but not simultaneous,
glucagon and cholecystokinin infusions inhibit feeding in men. Am J Physiol Regul Integr 289. Grandt D, Schimiczek M, Struk K, Shively J, Eysselein VE, Goebell H, Reeve JR Jr.
Comp Physiol 262: R975–R980, 1992. Characterization of two forms of peptide YY, PYY(1–36) and PYY(3–36), in the
rabbit. Peptides 15: 815– 820, 1994.
270. Geary N, Moran TH. Basic science of appetite. In: Comprehensive Textbook of Psychi-
atry (9th ed.), edited by Sadock BJ, Sadock VA, Ruiz P.New York: Wolters Kluwer/ 290. Greenough A, Cole G, Lewis J, Lockton A, Blundell J. Untangling the effects of hunger,
Lippincott Williams & Wilkens, 2009, p. 375–387. anxiety, and nausea on energy intake during intravenous cholecystokinin octapeptide
(CCK-8) infusion. Physiol Behav 65: 303–310, 1998.
271. Geary N, Moran TH. Basic science of appetite. In: Comprehensive Textbook of Psychi-
atry (10th ed.), edited by Sadock BJ, Sadock VA, Ruiz P. New York: Wolters Kluwer/ 291. Grill HJ, Hayes MR. Hindbrain neurons as an essential hub in the neuroanatomically
Lippincott Williams & Wilkens. In press. distributed control of energy balance. Cell Metab 16: 296 –309, 2012.
272. Geeraerts B, Van Oudenhove L, Dupont P, Vanderghinste D, Bormans G, Van Laere 292. Grossman MI. Gastrointestinal hormones. Physiol Rev 30: 33–90, 1950.
K, Tack J. Different regional brain activity during physiological gastric distension com-
pared to balloon distension: a H2 15O-PET study. Neurogastroenterol Motil 23: 533– 293. Grossman MI. Physiological effects of gastrointestinal hormones. Federation Proc 36:
e203, 2011. 1930 –1932, 1977.
273. Geliebter A, Westreich S, Gage D. Gastric distention by balloon and test-meal intake 294. Grundy D. Signalling the state of the digestive tract. Auton Neurosci 125: 76 – 80,
in obese and lean subjects. Am J Clin Nutr 48: 592–594, 1988. 2006.
274. Gentilcore D, Bryant B, Wishart JM, Morris HA, Horowitz M, Jones KL. Acarbose 295. Gryback P, Naslund E, Hellstrom PM, Jacobsson H, Backman L. Gastric emptying of
attenuates the hypotensive response to sucrose and slows gastric emptying in the solids in humans: improved evaluation by Kaplan-Meier plots, with special reference
elderly. Am J Med 118: 1289, 2005. to obesity and gender. Eur J Nucl Med 23: 1562–1567, 1996.
275. Gerspach AC, Steinert RE, Schonenberger L, Graber-Maier A, Beglinger C. The role 296. Gu N, Tsuda M, Matsunaga T, Adachi T, Yasuda K, Ishihara A, Tsuda K. Glucose
of the gut sweet taste receptor in regulating GLP-1, PYY, and CCK release in humans. regulation of dipeptidyl peptidase IV gene expression is mediated by hepatocyte
Am J Physiol Endocrinol Metab 301: E317–E325, 2011. nuclear factor-1alpha in epithelial intestinal cells. Clin Exp Pharmacol Physiol 35: 1433–
1439, 2008.
276. Gibbons C, Caudwell P, Finlayson G, Webb DL, Hellstrom PM, Naslund E, Blundell JE.
Comparison of postprandial profiles of ghrelin, active GLP-1, and total PYY to meals 297. Gunnarsson PT, Winzell MS, Deacon CF, Larsen MO, Jelic K, Carr RD, Ahren B.
varying in fat and carbohydrate and their association with hunger and the phases of Glucose-induced incretin hormone release and inactivation are differently modulated
satiety. J Clin Endocrinol Metab 98: 847– 855, 2013. by oral fat and protein in mice. Endocrinology 147: 3173–3180, 2006.
277. Gibbons C, Finlayson G, Caudwell P, Webb DL, Hellstrom PM, Naslund E, Blundell JE. 298. Gutzwiller JP, Hruz P, Huber AR, Hamel C, Zehnder C, Drewe J, Gutmann H, Stanga
Postprandial profiles of CCK after high fat and high carbohydrate meals and the Z, Vogel D, Beglinger C. Glucagon-like peptide-1 is involved in sodium and water
relationship to satiety in humans. Peptides 77: 3– 8, 2016. homeostasis in humans. Digestion 73: 142–150, 2006.
299. Gutzwiller JP, Degen L, Matzinger D, Prestin S, Beglinger C. Interaction between 319. Helou N, Obeid O, Azar ST, Hwalla N. Variation of postprandial PYY 3–36 response
GLP-1 and CCK-33 in inhibiting food intake and appetite in men. Am J Physiol Regul following ingestion of differing macronutrient meals in obese females. Ann Nutr Metab
Integr Comp Physiol 287: R562–R567, 2004. 52: 188 –195, 2008.
300. Gutzwiller JP, Drewe J, Göke B, Schmidt H, Rohrer B, Lareida J, Beglinger C. Gluca- 320. Henderson J. Ernest Starling and ‘Hormones’: an historical commentary. J Endocrinol
gon-like peptide-1 promotes satiety and reduces food intake in patients with diabetes 184: 5–10, 2005.
mellitus type 2. Am J Physiol Regul Integr Comp Physiol 276: R1541–R1544, 1999.
321. Herman CP, Polivy J. Normative influences on food intake. Physiol Behav 86: 762–772,
301. Gutzwiller JP, Göke B, Drewe J, Hildebrand P, Ketterer S, Handschin D, Winterhalder 2005.
R, Conen D, Beglinger C. Glucagon-like peptide-1: a potent regulator of food intake
322. Herrmann C, Goke R, Richter G, Fehmann HC, Arnold R, Goke B. Glucagon-like
in humans. Gut 44: 81– 86, 1999. peptide-1 and glucose-dependent insulin-releasing polypeptide plasma levels in re-
sponse to nutrients. Digestion 56: 117–126, 1995.
302. Häberer D, Tasker M, Foltz M, Geary N, Westerterp M, Langhans W. Intragastric
infusion of pea-protein hydrolysate reduces test-meal size in rats more than pea 323. Hidalgo L, Clave P, Estorch M, Rodriguez-Espinosa J, Rovati L, Greeley GH, Capella G,
protein. Physiol Behav 104: 1041–1047, 2011. Lluis F. Effect of cholecystokinin-A receptor blockade on postprandial insulinaemia
and gastric emptying in humans. Neurogastroenterol Motil 14: 519 –525, 2002.
303. Habib AM, Richards P, Cairns LS, Rogers GJ, Bannon CA, Parker HE, Morley TC, Yeo
GS, Reimann F, Gribble FM. Overlap of endocrine hormone expression in the mouse 324. Hildebrand P, Ensinck JW, Ketterer S, Delco F, Mossi S, Bangerter U, Beglinger C.
intestine revealed by transcriptional profiling and flow cytometry. Endocrinology 153: Effect of a cholecystokinin antagonist on meal-stimulated insulin and pancreatic poly-
3054 –3065, 2012. peptide release in humans. J Clin Endocrinol Metab 72: 1123–1129, 1991.
304. Habib AM, Richards P, Rogers GJ, Reimann F, Gribble FM. Co-localisation and secre- 325. Hildebrand P, Petrig C, Burckhardt B, Ketterer S, Lengsfeld H, Fleury A, Hadvary P,
tion of glucagon-like peptide 1 and peptide YY from primary cultured human L cells. Beglinger C. Hydrolysis of dietary fat by pancreatic lipase stimulates cholecystokinin
Diabetologia 56: 1413–1416, 2013. release. Gastroenterology 114: 123–129, 1998.
305. Hajnal A, Kovacs P, Ahmed T, Meirelles K, Lynch CJ, Cooney RN. Gastric bypass 326. Hill BR, De Souza MJ, Wagstaff DA, Sato R, Williams NI. 24-hour profiles of circulating
surgery alters behavioral and neural taste functions for sweet taste in obese rats. Am ghrelin and peptide YY are inversely associated in normal weight premenopausal
J Physiol Gastrointest Liver Physiol 299: G967–G979, 2010. women. Peptides 38: 159 –162, 2012.
306. Halatchev IG, Cone RD. Peripheral administration of PYY(3–36) produces condi- 327. Himeno S, Tarui S, Kanayama S, Kuroshima T, Shinomura Y, Hayashi C, Tateishi K,
tioned taste aversion in mice. Cell Metab 1: 159 –168, 2005. Imagawa K, Hashimura E, Hamaoka T. Plasma cholecystokinin responses after inges-
tion of liquid meal and intraduodenal infusion of fat, amino acids, or hydrochloric acid
307. Hammer HF. Medical complications of bariatric surgery: focus on malabsorption and in man: analysis with region specific radioimmunoassay. Am J Gastroenterol 78: 703–
dumping syndrome. Dig Dis 30: 182–186, 2012. 707, 1983.
308. Hansen M, Hjollund KR, Hartmann B, Plamboeck A, Deacon CF, Wewer Albrechtsen 328. Hira T, Nakajima S, Eto Y, Hara H. Calcium-sensing receptor mediates phenylalanine-
NJ, Holst JJ. Important species differences regarding lymph contribution to gut hor- induced cholecystokinin secretion in enteroendocrine STC-1 cells. FEBS J 275: 4620 –
mone responses. Peptides 71: 28 –31, 2015. 4626, 2008.
312. Hayes MR, Bradley L, Grill HJ. Endogenous hindbrain glucagon-like peptide-1 recep- 332. Hojberg PV, Vilsboll T, Rabol R, Knop FK, Bache M, Krarup T, Holst JJ, Madsbad S.
tor activation contributes to the control of food intake by mediating gastric satiation Four weeks of near-normalisation of blood glucose improves the insulin response to
signaling. Endocrinology 150: 2654 –2659, 2009. glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide in patients
with type 2 diabetes. Diabetologia 52: 199 –207, 2009.
313. Hayes MR, Kanoski SE, De Jonghe BC, Leichner TM, Alhadeff AL, Fortin SM, Arnold
M, Langhans W, Grill HJ. The common hepatic branch of the vagus is not required to 333. Holdstock C, Zethelius B, Sundbom M, Karlsson FA, Eden Engstrom B. Postprandial
mediate the glycemic and food intake suppressive effects of glucagon-like-peptide-1. changes in gut regulatory peptides in gastric bypass patients. Int J Obes 32: 1640 –1646,
Am J Physiol Regul Integr Comp Physiol 301: R1479 –R1485, 2011. 2008.
314. Heading RC, Tothill P, McLoughlin GP, Shearman DJ. Gastric emptying rate measure- 334. Holdsworth CD, Dawson AM. The absorption of monosaccharides in man. Clin Sci 27:
ment in man. A double isotope scanning technique for simultaneous study of liquid and 371–379, 1964.
solid components of a meal. Gastroenterology 71: 45–50, 1976.
335. Holst JJ. Incretin hormones and the satiation signal. Int J Obes (Lond) 37: 1161–1168,
315. Heath RB, Jones R, Frayn KN, Robertson MD. Vagal stimulation exaggerates the 2013.
inhibitory ghrelin response to oral fat in humans. J Endocrinol 180: 273–281, 2004.
336. Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev 87: 1409 –1439, 2007.
316. Heddle R, Dent J, Read NW, Houghton LA, Toouli J, Horowitz M, Maddern GJ, 337. Hopman WP, Jansen JB, Lamers CB. Comparative study of the effects of equal
Downton J. Antropyloroduodenal motor responses to intraduodenal lipid infusion in amounts of fat, protein, and starch on plasma cholecystokinin in man. Scand J Gastro-
healthy volunteers. Am J Physiol Gastrointest Liver Physiol 254: G671–G679, 1988. enterol 20: 843– 847, 1985.
317. Heden TD, Liu Y, Sims L, Kearney ML, Whaley-Connell AT, Chockalingam A, Dell- 338. Horowitz M, Collins PJ, Shearman DJ. Effect of increasing the caloric/osmotic content
sperger KC, Fairchild TJ, Kanaley JA. Liquid meal composition, postprandial satiety of the liquid component of a mixed solid and liquid meal on gastric emptying in obese
hormones, and perceived appetite and satiety in obese women during acute caloric subjects. Hum Nutr Clin Nutr 40: 51–56, 1986.
restriction. Eur J Endocrinol 168: 593– 600, 2013.
339. Horowitz M, Cook DJ, Collins PJ, Harding PE, Hooper MJ, Walsh JF, Shearman DJ.
318. Hellstrom PM, Gryback P, Jacobsson H. The physiology of gastric emptying. Best Pract Measurement of gastric emptying after gastric bypass surgery using radionuclides. Br
Res Clin Anaesthesiol 20: 397– 407, 2006. J Surg 69: 655– 657, 1982.
340. Horowitz M, Cunningham KM, Wishart JM, Jones KL, Read NW. The effect of short- 360. Irwin N, Pathak V, Flatt PR. A novel CCK-8/GLP-1 hybrid peptide exhibiting promi-
term dietary supplementation with glucose on gastric emptying of glucose and fruc- nent insulinotropic, glucose-lowering, and satiety actions with significant therapeutic
tose and oral glucose tolerance in normal subjects. Diabetologia 39: 481– 486, 1996. potential in high-fat-fed mice. Diabetes 64: 2996 –3009, 2015.
341. Horowitz M, Dent J. Disordered gastric emptying: mechanical basis, assessment and 361. Isbell JM, Tamboli RA, Hansen EN, Saliba J, Dunn JP, Phillips SE, Marks-Shulman PA,
treatment. Baillieres Clin Gastroenterol 5: 371– 407, 1991. Abumrad NN. The importance of caloric restriction in the early improvements in insulin
sensitivity after Roux-en-Y gastric bypass surgery. Diabetes Care 33: 1438–1442, 2010.
342. Horowitz M, Edelbroek MA, Wishart JM, Straathof JW. Relationship between oral
glucose tolerance and gastric emptying in normal healthy subjects. Diabetologia 36: 362. Ivy AC, Oldberg E. A hormone mechanism for gallbladder contraction and evacuation.
857– 862, 1993. Am J Physiol 65: 599 – 613, 1928.
343. Horowitz M, Flint A, Jones KL, Hindsberger C, Rasmussen MF, Kapitza C, Doran S, Jax 363. Jackness C, Karmally W, Febres G, Conwell IM, Ahmed L, Bessler M, McMahon DJ,
T, Zdravkovic M, Chapman IM. Effect of the once-daily human GLP-1 analogue Korner J. Very low-calorie diet mimics the early beneficial effect of Roux-en-Y gastric
liraglutide on appetite, energy intake, energy expenditure and gastric emptying in type bypass on insulin sensitivity and beta-cell function in type 2 diabetic patients. Diabetes
2 diabetes. Diabetes Res Clin Pract 97: 258 –266, 2012. 62: 3027–3032, 2013.
344. Horowitz M, Harding PE, Maddox AF, Wishart JM, Akkermans LM, Chatterton BE, 364. Jacobsen SH, Olesen SC, Dirksen C, Jorgensen NB, Bojsen-Moller KN, Kielgast U,
Worm D, Almdal T, Naver LS, Hvolris LE, Rehfeld JF, Wulff BS, Clausen TR, Hansen
Shearman DJ. Gastric and oesophageal emptying in patients with type 2 (non-insulin-
DL, Holst JJ, Madsbad S. Changes in gastrointestinal hormone responses, insulin
dependent) diabetes mellitus. Diabetologia 32: 151–159, 1989.
sensitivity, and beta-cell function within 2 weeks after gastric bypass in non-diabetic
345. Horowitz M, Maddox A, Bochner M, Wishart J, Bratasiuk R, Collins P, Shearman D. subjects. Obes Surg 22: 1084 –1096, 2012.
Relationships between gastric emptying of solid and caloric liquid meals and alcohol
365. Jang HJ, Kokrashvili Z, Theodorakis MJ, Carlson OD, Kim BJ, Zhou J, Kim HH, Xu X,
absorption. Am J Physiol Gastrointest Liver Physiol 257: G291–G298, 1989.
Chan SL, Juhaszova M, Bernier M, Mosinger B, Margolskee RF, Egan JM. Gut-ex-
346. Hosoda H, Kangawa K. The autonomic nervous system regulates gastric ghrelin se- pressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1.
cretion in rats. Regul Pept 146: 12–18, 2008. Proc Natl Acad Sci USA 104: 15069 –15074, 2007.
347. Hou Q, Lin Z, Dusing RW, Gajewski BJ, McCallum RW. A Bayesian hierarchical 366. Janssen P, Vanden Berghe P, Verschueren S, Lehmann A, Depoortere I, Tack J.
Review article: the role of gastric motility in the control of food intake. Aliment
assessment of gastric emptying with the linear, power exponential and modified
Pharmacol Ther 33: 880 – 894, 2011.
power exponential models. Neurogastroenterol Motil 22: 1308 –1317, 2010.
367. Janssen S, Laermans J, Verhulst Pj Thijs T, Tack J, Depoortere I. Bitter taste receptors
348. Howard AD, Feighner SD, Cully DF, Arena JP, Liberator PA, Rosenblum CI, Hamelin
and alpha-gustducin regulate the secretion of ghrelin with functional effects on food
M, Hreniuk DL, Palyha OC, Anderson J, Paress PS, Diaz C, Chou M, Liu KK, McKee
intake and gastric emptying. Proc Natl Acad Sci USA 108: 2094 –2099, 2011.
KK, Pong SS, Chaung LY, Elbrecht A, Dashkevicz M, Heavens R, Rigby M, Sirinathsing-
hji DJ, Dean DC, Melillo DG, Patchett AA, Nargund R, Griffin PR, DeMartino JA, 368. Jeffery RW, Harnack LJ. Evidence implicating eating as a primary driver for the obesity
Gupta SK, Schaeffer JM, Smith RG, Van der Ploeg LH. A receptor in pituitary and epidemic. Diabetes 56: 2673–2676, 2007.
hypothalamus that functions in growth hormone release. Science 273: 974 –977, 1996.
369. Jenkins AL, Jenkins DJ, Zdravkovic U, Wursch P, Vuksan V. Depression of the glycemic
349. Hunt JN, Stubbs DF. The volume and energy content of meals as determinants of index by high levels of beta-glucan fiber in two functional foods tested in type 2
gastric emptying. J Physiol 245: 209 –225, 1975. diabetes. Eur J Clin Nutr 56: 622– 628, 2002.
350. Hutson WR, Roehrkasse RL, Wald A. Influence of gender and menopause on gastric 370. Jerlhag E, Janson AC, Waters S, Engel JA. Concomitant release of ventral tegmental
emptying and motility. Gastroenterology 96: 11–17, 1989. acetylcholine and accumbal dopamine by ghrelin in rats. PLoS One 7: e49557, 2012.
351. Hveem K, Jones KL, Chatterton BE, Horowitz M. Scintigraphic measurement of 371. Jimenez A, Casamitjana R, Viaplana-Masclans J, Lacy A, Vidal J. GLP-1 action and
gastric emptying and ultrasonographic assessment of antral area: relation to appetite. glucose tolerance in subjects with remission of type 2 diabetes after gastric bypass
Gut 38: 816 – 821, 1996. surgery. Diabetes Care 36: 2062–2069, 2013.
352. Hvidberg A, Nielsen MT, Hilsted J, Orskov C, Holst JJ. Effect of glucagon-like pep- 372. Jonderko K. Comparative analysis of quantitative gastric emptying indices and power-
tide-1 (proglucagon 78-107amide) on hepatic glucose production in healthy man. exponential modelling of gastric emptying curves. Clin Phys Physiol Meas 10: 161–170,
Metabolism 43: 104 –108, 1994. 1989.
353. Ikramuddin S, Blackstone RP, Brancatisano A, Toouli J, Shah SN, Wolfe BM, Fujioka K, 373. Jones KL, Doran SM, Hveem K, Bartholomeusz FD, Morley JE, Sun WM, Chatterton
Maher JW, Swain J, Que FG, Morton JM, Leslie DB, Brancatisano R, Kow L, O’Rourke BE, Horowitz M. Relation between postprandial satiation and antral area in normal
RW, Deveney C, Takata M, Miller CJ, Knudson MB, Tweden KS, Shikora SA, Sarr MG, subjects. Am J Clin Nutr 66: 127–132, 1997.
Billington CJ. Effect of reversible intermittent intra-abdominal vagal nerve blockade
374. Jones RB, McKie S, Astbury N, Little TJ, Tivey S, Lassman DJ, McLaughlin J, Luckman
on morbid obesity: the ReCharge randomized clinical trial. JAMA 312: 915–922, 2014.
S, Williams SR, Dockray GJ, Thompson DG. Functional neuroimaging demonstrates
354. Imeryuz N, Yegen BC, Bozkurt A, Coskun T, Villanueva-Penacarrillo ML, Ulusoy NB. that ghrelin inhibits the central nervous system response to ingested lipid. Gut 61:
Glucagon-like peptide-1 inhibits gastric emptying via vagal afferent-mediated central 1543–1551, 2012.
mechanisms. Am J Physiol Gastrointest Liver Physiol 273: G920 –G927, 1997. 375. Jorgensen NB, Dirksen C, Bojsen-Moller KN, Jacobsen SH, Worm D, Hansen DL, Kris-
tiansen VB, Naver L, Madsbad S, Holst JJ. Exaggerated glucagon-like peptide 1 response is
355. Ionut V, Hucking K, Liberty IF, Bergman RN. Synergistic effect of portal glucose and
important for improved beta-cell function and glucose tolerance after Roux-en-Y gastric by-
glucagon-like peptide-1 to lower systemic glucose and stimulate counter-regulatory
pass in patients with type 2 diabetes. Diabetes 62: 3044–3052, 2013.
hormones. Diabetologia 48: 967–975, 2005.
376. Kaji I, Karaki S, Kuwahara A. Short-chain fatty acid receptor and its contribution to
357. Irwin N, Frizelle P, Montgomery IA, Moffett RC, O’Harte FP, Flatt PR. Beneficial
glucagon-like peptide-1 release. Digestion 89: 31–36, 2014.
effects of the novel cholecystokinin agonist (pGlu-Gln)-CCK-8 in mouse models of
obesity/diabetes. Diabetologia 55: 2747–2758, 2012. 377. Kalarchian MA, Marcus MD, Courcoulas AP, Cheng Y, Levine MD. Self-report of gastrointes-
tinal side effects after bariatric surgery. Surg Obes Relat Dis 10: 1202–1207, 2014.
358. Irwin N, Montgomery IA, Flatt PR. Comparison of the metabolic effects of sustained
CCK1 receptor activation alone and in combination with upregulated leptin signalling 378. Kanoski SE, Hayes MR, Skibicka KP. GLP-1 and weight loss: unraveling the diverse
in high-fat-fed mice. Diabetologia 56: 1425–1435, 2013. neural circuitry. Am J Physiol Regul Integr Comp Physiol 310: R885–R895, 2016.
359. Irwin N, Montgomery IA, O’Harte FP, Frizelle P, Flatt PR. Comparison of the inde- 379. Kanoski SE, Rupprecht LE, Fortin SM, De Jonghe BC, Hayes MR. The role of nausea in
pendent and combined metabolic effects of subchronic modulation of CCK and GIP food intake and body weight suppression by peripheral GLP-1 receptor agonists,
receptor action in obesity-related diabetes. Int J Obes (Lond) 37: 1058 –1063, 2013. exendin-4 and liraglutide. Neuropharmacology 62: 1916 –1927, 2012.
380. Karaki S, Mitsui R, Hayashi H, Kato I, Sugiya H, Iwanaga T, Furness JB, Kuwahara A. of food intake yet is not essential for the maintenance of body weight. J Clin Invest 103:
Short-chain fatty acid receptor, GPR43, is expressed by enteroendocrine cells and 383–391, 1999.
mucosal mast cells in rat intestine. Cell Tissue Res 324: 353–360, 2006.
403. Korner J, Inabnet W, Febres G, Conwell IM, McMahon DJ, Salas R, Taveras C, Schrope
381. Kastin AJ, Akerstrom V, Pan W. Interactions of glucagon-like peptide-1 (GLP-1) with B, Bessler M. Prospective study of gut hormone and metabolic changes after adjust-
the blood-brain barrier. J Mol Neurosci 18: 7–14, 2002. able gastric banding and Roux-en-Y gastric bypass. Int J Obes (Lond) 33: 786 –795,
2009.
382. Katschinski M, Schirra J, Begliner C, Langbein S, Wank U, D’Amato M, Arnold R.
Intestinal phase of human antro-pyloro-duodenal motility: cholinergic and CCK-me- 404. Kost LJ, Gores GJ, Sayles JM, Miller LJ, Lemasters JJ, Herman B, LaRusso NF. Lack of
diated regulation. Eur J Clin Invest 26: 574 –583, 1996. metabolic effects of cholecystokinin on hepatocytes. Hepatology 12: 301–305, 1990.
383. Kellum JM, Kuemmerle JF, O’Dorisio TM, Rayford P, Martin D, Engle K, Wolf L, 405. Kral JG, Paez W, Wolfe BM. Vagal nerve function in obesity: therapeutic implications.
Sugerman HJ. Gastrointestinal hormone responses to meals before and after gastric World J Surg 33: 1995–2006, 2009.
bypass and vertical banded gastroplasty. Ann Surg 211: 763–770, 1990.
406. Kreymann B, Williams G, Ghatei MA, Bloom SR. Glucagon-like peptide-1 7–36: a
384. Kent DM, Rothwell PM, Ioannidis JP, Altman DG, Hayward RA. Assessing and report- physiological incretin in man. Lancet 2: 1300 –1304, 1987.
ing heterogeneity in treatment effects in clinical trials: a proposal. Trials 11: 85, 2010.
407. Krieger JP, Arnold M, Pettersen KG, Lossel P, Langhans W, Lee SJ. Knockdown of
385. Kim BJ, Carlson OD, Jang HJ, Elahi D, Berry C, Egan JM. Peptide YY is secreted after GLP-1 receptors in vagal afferents affects normal food intake and glycemia. Diabetes
oral glucose administration in a gender-specific manner. J Clin Endocrinol Metab 90: 65: 34 – 43, 2016.
6665– 6671, 2005.
408. Kruszynska YT, Home PD, Hanning I, Alberti KG. Basal and 24-h C-peptide and
387. Kim DH, D’Alessio DA, Woods SC, Seeley RJ. The effects of GLP-1 infusion in the insulin secretion rate in normal man. Diabetologia 30: 16 –21, 1987.
hepatic portal region on food intake. Regul Pept 155: 110 –114, 2009.
409. Kuhre RE, Gribble FM, Hartmann B, Reimann F, Windelov JA, Rehfeld JF, Holst JJ.
388. Kinzig KP, D’Alessio DA, Seeley RJ. The diverse roles of specific GLP-1 receptors in Fructose stimulates GLP-1 but not GIP secretion in mice, rats, and humans. Am J
the control of food intake and the response to visceral illness. J Neurosci 22: 10470 – Physiol Gastrointest Liver Physiol 306: G622–G630, 2014.
10476, 2002.
410. Kuhre RE, Wewer Albrechtsen NJ, Hartmann B, Deacon CF, Holst JJ. Measurement
389. Kirchner H, Gutierrez JA, Solenberg PJ, Pfluger PT, Czyzyk TA, Willency JA, Schur- of the incretin hormones: glucagon-like peptide-1 and glucose-dependent insulino-
mann A, Joost HG, Jandacek RJ, Hale JE, Heiman ML, Tschop MH. GOAT links dietary tropic peptide. J Diabetes Complications 29: 445– 450, 2015.
lipids with the endocrine control of energy balance. Nat Med 15: 741–745, 2009.
411. Kuo P, Bellon M, Wishart J, Smout AJ, Holloway RH, Fraser RJ, Horowitz M, Jones KL,
390. Kirchner H, Heppner KM, Tschop MH. The role of ghrelin in the control of energy Rayner CK. Effects of metoclopramide on duodenal motility and flow events, glucose
balance. Handb Exp Pharmacol 161–184, 2012. absorption, and incretin hormone release in response to intraduodenal glucose infu-
sion. Am J Physiol Gastrointest Liver Physiol 299: G1326 –G1333, 2010.
391. Kissileff HR, Pi-Sunyer FX, Thornton J, Smith GP. C-terminal octapeptide of chole-
cystokinin decreases food intake in man. Am J Clin Nutr 34: 154 –160, 1981. 412. Kuo P, Chaikomin R, Pilichiewicz A, O’Donovan D, Wishart JM, Meyer JH, Jones KL,
Feinle-Bisset C, Horowitz M, Rayner CK. Transient, early release of glucagon-like
392. Kjems LL, Holst JJ, Volund A, Madsbad S. The influence of GLP-1 on glucose-stimu-
lated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic sub- peptide-1 during low rates of intraduodenal glucose delivery. Regul Peptides 146: 1–3,
jects. Diabetes 52: 380 –386, 2003. 2008.
393. Knauf C, Cani PD, Perrin C, Iglesias MA, Maury JF, Bernard E, Benhamed F, Gre- 413. Kurashina T, Dezaki K, Yoshida M, Sukma Rita R, Ito K, Taguchi M, Miura R, Tominaga
meaux T, Drucker DJ, Kahn CR, Girard J, Tanti JF, Delzenne NM, Postic C, Burcelin M, Ishibashi S, Kakei M, Yada T. The beta-cell GHSR and downstream cAMP/TRPM2
R. Brain glucagon-like peptide-1 increases insulin secretion and muscle insulin resis- signaling account for insulinostatic and glycemic effects of ghrelin. Sci Rep 5: 14041,
tance to favor hepatic glycogen storage. J Clin Invest 115: 3554 –3563, 2005. 2015.
394. Knight LC, Parkman HP, Brown KL, Miller MA, Trate DM, Maurer AH, Fisher RS. 414. Ladenheim EE. Liraglutide and obesity: a review of the data so far. Drug Des Dev Ther
Delayed gastric emptying and decreased antral contractility in normal premenopausal 9: 1867–1875, 2015.
women compared with men. Am J Gastroenterol 92: 968 –975, 1997.
415. Laferrere B, Teixeira J, McGinty J, Tran H, Egger JR, Colarusso A, Kovack B, Bawa B,
395. Koda S, Date Y, Murakami N, Shimbara T, Hanada T, Toshinai K, Niijima A, Furuya M, Koshy N, Lee H, Yapp K, Olivan B. Effect of weight loss by gastric bypass surgery
Inomata N, Osuye K, Nakazato M. The role of the vagal nerve in peripheral PYY3- versus hypocaloric diet on glucose and incretin levels in patients with type 2 diabetes.
36-induced feeding reduction in rats. Endocrinology 146: 2369 –2375, 2005. J Clin Endocrinol Metab 93: 2479 –2485, 2008.
396. Kohli R, Bradley D, Setchell KD, Eagon JC, Abumrad N, Klein S. Weight loss induced 416. Lamberts SW, Romijn JA, Wiersinga WM. The future endocrine patient. Reflections
by Roux-en-Y gastric bypass but not laparoscopic adjustable gastric banding increases on the future of clinical endocrinology. Eur J Endocrinol 149: 169 –175, 2003.
circulating bile acids. J Clin Endocrinol Metab 98: E708 –712, 2013.
417. Lamont BJ, Li Y, Kwan E, Brown TJ, Gaisano H, Drucker DJ. Pancreatic GLP-1
397. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a receptor activation is sufficient for incretin control of glucose metabolism in mice. J
growth-hormone-releasing acylated peptide from stomach. Nature 402: 656 – 660, Clin Invest 122: 388 – 402, 2012.
1999.
418. Larsson LI, Goltermann N, de Magistris L, Rehfeld JF, Schwartz TW. Somatostatin cell
398. Kojima M, Kangawa K. Ghrelin: structure and function. Physiol Rev 85: 495–522, 2005. processes as pathways for paracrine secretion. Science 205: 1393–1395, 1979.
399. Kong MF, Chapman I, Goble E, Wishart J, Wittert G, Morris H, Horowitz M. Effects of 419. Lassman DJ, McKie S, Gregory LJ, Lal S, D’Amato M, Steele I, Varro A, Dockray GJ,
oral fructose and glucose on plasma GLP-1 and appetite in normal subjects. Peptides Williams SC, Thompson DG. Defining the role of cholecystokinin in the lipid-induced
20: 545–551, 1999. human brain activation matrix. Gastroenterology 138: 1514 –1524, 2010.
400. Konturek JW, Stoll R, Gutwinska-Konturek M, Konturek SJ, Domschke W. Chole- 420. Lauffer LM, Iakoubov R, Brubaker PL. GPR119 is essential for oleoylethanolamide-
cystokinin in the regulation of gastric acid and endocrine pancreatic secretion in induced glucagon-like peptide-1 secretion from the intestinal enteroendocrine L-cell.
humans. Scand J Gastroenterol 28: 401– 407, 1993. Diabetes 58: 1058 –1066, 2009.
401. Konturek PC, Konturek SJ. The history of gastrointestinal hormones and the Polish 421. Laurenius A, Larsson I, Bueter M, Melanson KJ, Bosaeus I, Forslund HB, Lonroth H,
contribution to elucidation of their biology and relation to nervous system. J Physiol Fandriks L, Olbers T. Changes in eating behaviour and meal pattern following Roux-
Pharmacol 54 Suppl 3: 83–98, 2003. en-Y gastric bypass. Int J Obes (Lond) 36: 348 –355, 2012.
402. Kopin AS, Mathes WF, McBride EW, Nguyen M, Al-Haider W, Schmitz F, Bonner- 422. Le Roux CW, Aylwin SJ, Batterham RL, Borg CM, Coyle F, Prasad V, Shurey S, Ghatei
Weir S, Kanarek R, Beinborn M. The cholecystokinin-A receptor mediates inhibition MA, Patel AG, Bloom SR. Gut hormone profiles following bariatric surgery favor an
anorectic state, facilitate weight loss, and improve metabolic parameters. Ann Surg 443. Liou AP, Lu X, Sei Y, Zhao X, Pechhold S, Carrero RJ, Raybould HE, Wank S. The
243: 108 –114, 2006. G-protein-coupled receptor GPR40 directly mediates long-chain fatty acid-induced
secretion of cholecystokinin. Gastroenterology 140: 903–912, 2011.
423. Le Roux CW, Batterham RL, Aylwin SJ, Patterson M, Borg CM, Wynne KJ, Kent A,
Vincent RP, Gardiner J, Ghatei MA, Bloom SR. Attenuated peptide YY release in obese 444. Lippl F, Erdmann J, Steiger A, Lichter N, Czogalla-Peter C, Bidlingmaier M, Tholl S,
subjects is associated with reduced satiety. Endocrinology 147: 3– 8, 2006. Schusdziarra V. Low-dose ghrelin infusion– evidence against a hormonal role in food
intake. Regul Pept 174: 26 –31, 2012.
424. Le Roux CW, Bueter M, Theis N, Werling M, Ashrafian H, Lowenstein C, Atha-
nasiou T, Bloom SR, Spector AC, Olbers T, Lutz TA. Gastric bypass reduces fat 445. Little TJ, Doran S, Meyer JH, Smout AJ, O’Donovan DG, Wu KL, Jones KL, Wishart J,
intake and preference. Am J Physiol Regul Integr Comp Physiol 301: R1057–R1066, Rayner CK, Horowitz M, Feinle-Bisset C. The release of GLP-1 and ghrelin, but not
2011. GIP and CCK, by glucose is dependent upon the length of small intestine exposed. Am
J Physiol Endocrinol Metab 291: E647–E655, 2006.
425. Le Roux CW, Welbourn R, Werling M, Osborne A, Kokkinos A, Laurenius A, Lonroth
H, Fandriks L, Ghatei MA, Bloom SR, Olbers T. Gut hormones as mediators of 446. Little TJ, Feltrin KL, Horowitz M, Smout AJ, Rades T, Meyer JH, Pilichiewicz AN,
appetite and weight loss after Roux-en-Y gastric bypass. Ann Surg 246: 780 –785, 2007. Wishart J, Feinle-Bisset C. Dose-related effects of lauric acid on antropyloroduodenal
motility, gastrointestinal hormone release, appetite, and energy intake in healthy men.
426. Le Sauter J, Goldberg B, Geary N. CCK inhibits real and sham feeding in gastric Am J Physiol Regul Integr Comp Physiol 289: R1090 –R1098, 2005.
vagotomized rats. Physiol Behav 44: 527–534, 1988.
447. Little TJ, Gopinath A, Patel E, McGlone A, Lassman DJ, D’Amato M, McLaughlin JT,
427. Lejeune MP, Westerterp KR, Adam TC, Luscombe-Marsh ND, Westerterp-Plant- Thompson DG. Gastric emptying of hexose sugars: role of osmolality, molecular
enga MS. Ghrelin and glucagon-like peptide 1 concentrations, 24-h satiety, and energy structure and the CCK1 receptor. Neurogastroenterol Motil 22: 1183–1190, 2010.
and substrate metabolism during a high-protein diet and measured in a respiration
chamber. Am J Clin Nutr 83: 89 –94, 2006. 448. Little TJ, Pilichiewicz AN, Russo A, Phillips L, Jones KL, Nauck MA, Wishart J,
Horowitz M, Feinle-Bisset C. Effects of intravenous glucagon-like peptide-1 on
428. Lepage R, Albert C. Fifty years of development in the endocrinology laboratory. Clin gastric emptying and intragastric distribution in healthy subjects: relationships
Biochem 39: 542–557, 2006. with postprandial glycemic and insulinemic responses. J Clin Endocrinol Metab 91:
1916 –1923, 2006.
429. Levin F, Edholm T, Schmidt PT, Gryback P, Jacobsson H, Degerblad M, Hoybye C,
Holst JJ, Rehfeld JF, Hellstrom PM, Naslund E. Ghrelin stimulates gastric emptying and 449. Liu EH, Oberg K. The history and development of the gastroenteropancreatic endo-
hunger in normal-weight humans. J Clin Endocrinol Metab 91: 3296 –3302, 2006. crine axis. Endocrinol Metab Clin North Am 39: 697–711, 2010.
430. Li P, Tiwari HK, Lin WY, Allison DB, Chung WK, Leibel RL, Yi N, Liu N. Genetic 450. Llewellyn-Smith IJ, Reimann F, Gribble FM, Trapp S. Preproglucagon neurons project
association analysis of 30 genes related to obesity in a European American population. widely to autonomic control areas in the mouse brain. Neuroscience 180: 111–121,
Int J Obes (Lond) 38: 724 –729, 2014. 2011.
431. Liddle RA. Cholecystokinin cells. Annu Rev Physiol 59: 221–242, 1997. 451. Lo CM, Xu M, Yang Q, Zheng S, Carey KM, Tubb MR, Davidson WS, Liu M, Woods
432. Liddle RA. Regulation of cholecystokinin secretion by intraluminal releasing factors. SC, Tso P. Effect of intraperitoneal and intravenous administration of cholecysto-
Am J Physiol Gastrointest Liver Physiol 269: G319 –G327, 1995. kinin-8 and apolipoprotein AIV on intestinal lymphatic CCK-8 and apo AIV concen-
tration. Am J Physiol Regul Integr Comp Physiol 296: R43–R50, 2009.
433. Liddle RA, Gertz BJ, Kanayama S, Beccaria L, Coker LD, Turnbull TA, Morita ET.
Effects of a novel cholecystokinin (CCK) receptor antagonist, MK-329, on gallbladder 452. Locatelli I, Mrhar A, Bogataj M. Gastric emptying of pellets under fasting conditions: a
contraction and gastric emptying in humans. Implications for the physiology of CCK. mathematical model. Pharm Res 26: 1607–1617, 2009.
J Clin Invest 84: 1220 –1225, 1989.
453. Lomenick JP, Clasey JL, Anderson JW. Meal-related changes in ghrelin, peptide YY,
434. Liddle RA, Gertz BJ, Kanayama S, Beccaria L, Gettys TW, Taylor IL, Rushakoff RJ, and appetite in normal weight and overweight children. Obesity 16: 547–552, 2008.
Williams VC, Coker LD. Regulation of pancreatic endocrine function by cholecysto-
454. Long SJ, Sutton Ja Amaee WB, Giouvanoudi a Spyrou NM, Rogers PJ, Morgan LM. No
kinin: studies with MK-329, a nonpeptide cholecystokinin receptor antagonist. J Clin
effect of glucagon-like peptide-1 on short-term satiety and energy intake in man. Br J
Endocrinol Metab 70: 1312–1318, 1990.
Nutr 81: 273–279, 1999.
435. Liddle RA, Goldfine ID, Rosen MS, Taplitz RA, Williams JA. Cholecystokinin bioactivity
455. Loop MS, Frazier-Wood AC, Thomas AS, Dhurandhar EJ, Shikany JM, Gadbury GL,
in human plasma. Molecular forms, responses to feeding, and relationship to gallblad-
Allison DB. Submitted for your consideration: potential advantages of a novel clinical
der contraction. J Clin Invest 75: 1144 –1152, 1985.
trial design and initial patient reaction. Front Genet 3: 145, 2012.
436. Liddle RA, Morita ET, Conrad CK, Williams JA. Regulation of gastric emptying in
456. Lu L, Louie D, Owyang C. A cholecystokinin releasing peptide mediates feedback
humans by cholecystokinin. J Clin Invest 77: 992–996, 1986.
regulation of pancreatic secretion. Am J Physiol Gastrointest Liver Physiol 256: G430 –
437. Liddle RA, Rushakoff RJ, Morita ET, Beccaria L, Carter JD, Goldfine ID. Physiological G435, 1989.
role for cholecystokinin in reducing postprandial hyperglycemia in humans. J Clin
457. Lu X, Zhao X, Feng J, Liou AP, Anthony S, Pechhold S, Sun Y, Lu H, Wank S.
Invest 81: 1675–1681, 1988.
Postprandial inhibition of gastric ghrelin secretion by long-chain fatty acid through
438. Lieverse RJ, Jansen JB, Masclee AA, Lamers CB. Satiety effects of a physiological dose GPR120 in isolated gastric ghrelin cells and mice. Am J Physiol Gastrointest Liver Physiol
of cholecystokinin in humans. Gut 36: 176 –179, 1995. 303: G367–G376, 2012.
439. Lieverse RJ, Jansen JB, Masclee AA, Rovati LC, Lamers CB. Effect of a low dose of 458. Lugari R, Dei Cas A, Ugolotti D, Barilli AL, Camellini C, Ganzerla GC, Luciani A,
intraduodenal fat on satiety in humans: studies using the type A cholecystokinin re- Salerni B, Mittenperger F, Nodari S, Gnudi A, Zandomeneghi R. Glucagon-like peptide
ceptor antagonist loxiglumide. Gut 35: 501–505, 1994. 1 (GLP-1) secretion and plasma dipeptidyl peptidase IV (DPP-IV) activity in morbidly
obese patients undergoing biliopancreatic diversion. Horm Metab Res 36: 111–115,
440. Lieverse RJ, Jansen JB, van de Zwan A, Samson L, Masclee AA, Lamers CB. Effects of 2004.
a physiological dose of cholecystokinin on food intake and postprandial satiation in
man. Regul Pept 43: 83– 89, 1993. 459. Ma J, Checklin HL, Wishart JM, Stevens JE, Jones KL, Horowitz M, Meyer JH, Rayner
CK. A randomised trial of enteric-coated nutrient pellets to stimulate gastrointestinal
441. Lin S, Boey D, Herzog H. NPY and Y receptors: lessons from transgenic and knockout peptide release and lower glycaemia in type 2 diabetes. Diabetologia 56: 1236 –1242,
models. Neuropeptides 38: 189 –200, 2004. 2013.
442. Lindgren O, Carr RD, Holst JJ, Deacon CF, Ahren B. Dissociated incretin hormone 460. Ma J, Pilichiewicz AN, Feinle-Bisset C, Wishart JM, Jones KL, Horowitz M, Rayner CK.
response to protein versus fat ingestion in obese subjects. Diabetes Obes Metab 13: Effects of variations in duodenal glucose load on glycaemic, insulin, and incretin re-
863– 865, 2011. sponses in type 2 diabetes. Diabet Med 29: 604 – 608, 2012.
461. Mace OJ, Schindler M, Patel S. The regulation of K- and L-cell activity by GLUT2 and 481. Maurer AH. Advancing gastric emptying studies: standardization and new parameters
the calcium-sensing receptor CasR in rat small intestine. J Physiol 590: 2917–2936, to assess gastric motility and function. Semin Nucl Med 42: 101–112, 2012.
2012.
482. Mayer J, Thomas DW. Regulation of food intake and obesity. Science 156: 328 –337,
462. MacIntosh CG, Andrews JM, Jones KL, Wishart JM, Morris HA, Jansen JB, Morley JE, 1967.
Horowitz M, Chapman IM. Effects of age on concentrations of plasma cholecysto-
kinin, glucagon-like peptide 1, and peptide YY and their relation to appetite and 483. Mayer LE, Schebendach J, Bodell LP, Shingleton RM, Walsh BT. Eating behavior in
pyloric motility. Am J Clin Nutr 69: 999 –1006, 1999. anorexia nervosa: before and after treatment. Int J Eat Disord 45: 290 –293, 2012.
463. Makhlouf GM. Neural and hormonal regulation of function in the gut. Hosp Pract 25: 484. McFarlane MR, Brown MS, Goldstein JL, Zhao TJ. Induced ablation of ghrelin cells in
79 – 87, 1990. adult mice does not decrease food intake, body weight, or response to high-fat diet.
Cell Metab 20: 54 – 60, 2014.
464. Malagelada C, Accarino A, Molne L, Mendez S, Campos E, Gonzalez A, Malagelada JR,
Azpiroz F. Digestive, cognitive and hedonic responses to a meal. Neurogastroenterol 485. McKay NJ, Galante DL, Daniels D. Endogenous glucagon-like peptide-1 reduces
drinking behavior and is differentially engaged by water and food intakes in rats. J
Motil 27: 389 –396, 2015.
Neurosci 34: 16417–16423, 2014.
465. Malik S, McGlone F, Bedrossian D, Dagher A. Ghrelin modulates brain activity in areas
486. McLaughlin J, Grazia Luca M, Jones MN, D’Amato M, Dockray GJ, Thompson DG.
that control appetitive behavior. Cell Metab 7: 400 – 409, 2008.
Fatty acid chain length determines cholecystokinin secretion and effect on human
466. Maljaars PW, Symersky T, Kee BC, Haddeman E, Peters HP, Masclee AA. Effect of gastric motility. Gastroenterology 116: 46 –53, 1999.
ileal fat perfusion on satiety and hormone release in healthy volunteers. Int J Obes
487. McLaughlin JT, Lomax RB, Hall L, Dockray GJ, Thompson DG, Warhurst G. Fatty
(Lond) 32: 1633–1639, 2008.
acids stimulate cholecystokinin secretion via an acyl chain length-specific, Ca2⫹-de-
467. Manning S, Batterham RL. The role of gut hormone peptide yy in energy and glucose pendent mechanism in the enteroendocrine cell line STC-1. J Physiol 513: 11–18,
homeostasis: twelve years on. Annu Rev Physiol 76: 585– 608, 2014. 1998.
468. Manning S, Pucci A, Batterham RL. GLP-1: a mediator of the beneficial metabolic 488. Medeiros MD, Turner AJ. Processing and metabolism of peptide-YY: pivotal roles of
effects of bariatric surgery? Physiology 30: 50 – 62, 2015. dipeptidylpeptidase-IV, aminopeptidase-P, and endopeptidase-24.11. Endocrinology
134: 2088 –2094, 1994.
469. Manning S, Pucci A, Batterham RL. Roux-en-Y gastric bypass: effects on feeding
behavior and underlying mechanisms. J Clin Invest 125: 939 –948, 2015. 489. Medvei VC. A History of Endocrinology. Lancaster, PA: MTP Press, 1982.
470. Marathe CS, Horowitz M, Trahair LG, Wishart JM, Bound M, Lange K, Rayner CK, 490. Meier JJ, Nauck MA. Glucagon-like peptide 1(GLP-1) in biology and pathology. Dia-
Jones KL. Relationships of “early” and “late” glycemic responses with gastric emptying betes Metab Res Rev 21: 91–117, 2005.
during an oral glucose tolerance test. J Clin Endocrinol Metab 100: 3565–3571, 2015.
491. Meier JJ, Nauck MA. Is the diminished incretin effect in type 2 diabetes just an epi-
471. Marathe CS, Rayner CK, Bound M, Checklin H, Standfield S, Wishart J, Lange K, Jones phenomenon of impaired beta-cell function? Diabetes 59: 1117–1125, 2010.
KL, Horowitz M. Small intestinal glucose exposure determines the magnitude of the
492. Melhorn SJ, Tyagi V, Smeraglio A, Roth CL, Schur EA. Initial evidence that GLP-1
incretin effect in health and type 2 diabetes. Diabetes 63: 2668 –2675, 2014.
receptor blockade fails to suppress postprandial satiety or promote food intake in
humans. Appetite 82C: 85–90, 2014.
472. Marathe CS, Rayner CK, Jones KL, Horowitz M. Relationships between gastric emp-
tying, postprandial glycemia, and incretin hormones. Diabetes Care 36: 1396 –1405, 493. Mentis N, Vardarli I, Kothe LD, Holst JJ, Deacon CF, Theodorakis M, Meier JJ, Nauck
2013. MA. GIP does not potentiate the antidiabetic effects of GLP-1 in hyperglycemic
patients with type 2 diabetes. Diabetes 60: 1270 –1276, 2011.
473. Marchal-Victorion S, Vionnet N, Escrieut C, Dematos F, Dina C, Dufresne M, Vaysse
N, Pradayrol L, Froguel P, Fourmy D. Genetic, pharmacological and functional analysis 494. Merkestein M, Brans MA, Luijendijk MC, de Jong JW, Egecioglu E, Dickson SL, Adan
of cholecystokinin-1 and cholecystokinin-2 receptor polymorphism in type 2 diabetes RA. Ghrelin mediates anticipation to a palatable meal in rats. Obesity 20: 963–971,
and obese patients. Pharmacogenetics 12: 23–30, 2002. 2012.
474. Marciani L, Gowland PA, Spiller RC, Manoj P, Moore RJ, Young P, Fillery-Travis AJ. 495. Meyer BM, Werth BA, Beglinger C, Hildebrand P, Jansen JB, Zach D, Rovati LC,
Effect of meal viscosity and nutrients on satiety, intragastric dilution, and emptying Stalder GA. Role of cholecystokinin in regulation of gastrointestinal motor functions.
assessed by MRI. Am J Physiol Gastrointest Liver Physiol 280: G1227–G1233, 2001. Lancet 2: 12–15, 1989.
475. Marciani L, Hall N, Pritchard SE, Cox EF, Totman JJ, Lad M, Hoad CL, Foster TJ, 496. Meyer-Gerspach AC, Steinert RE, Keller S, Malarski A, Schulte FH, Beglinger C.
Gowland PA, Spiller RC. Preventing gastric sieving by blending a solid/water meal Effects of chenodeoxycholic acid on the secretion of gut peptides and fibroblast
enhances satiation in healthy humans. J Nutr 142: 1253–1258, 2012. growth factors in healthy humans. J Clin Endocrinol Metab 98: 3351–3358, 2013.
476. Mari A, Bagger JI, Ferrannini E, Holst JJ, Knop FK, Vilsboll T. Mechanisms of the 497. Meyer-Gerspach AC, Wolnerhanssen B, Beglinger B, Nessenius F, Napitupulu M,
incretin effect in subjects with normal glucose tolerance and patients with type 2 Schulte FH, Steinert RE, Beglinger C. Gastric and intestinal satiation in obese and
diabetes. PLoS One 8: e73154, 2013. normal weight healthy people. Physiol Behav 129: 265–271, 2014.
477. Mathes CM, Bueter M, Smith KR, Lutz TA, le Roux CW, Spector AC. Roux-en-Y 498. Michaliszyn SF, Lee S, Bacha F, Tfayli H, Farchoukh L, Mari A, Ferrannini E, Arslanian
gastric bypass in rats increases sucrose taste-related motivated behavior independent S. Differences in beta-cell function and insulin secretion in Black vs. White obese
of pharmacological GLP-1-receptor modulation. Am J Physiol Regul Integr Comp Physiol adolescents: do incretin hormones play a role? Pediatr Diabetes. In press.
302: R751–R767, 2012.
499. Mietlicki-Baase EG, Ortinski PI, Reiner DJ, Sinon CG, McCutcheon JE, Pierce RC,
478. Maton PN, Selden AC, Chadwick VS. Differential distribution of molecular forms of Roitman MF, Hayes MR. Glucagon-like peptide-1 receptor activation in the nucleus
cholecystokinin in human and porcine small intestinal mucosa. Regul Pept 8: 9 –19, accumbens core suppresses feeding by increasing glutamatergic AMPA/kainate signal-
1984. ing. J Neurosci 34: 6985– 6992, 2014.
479. Matzinger D, Degen L, Drewe J, Meuli J, Duebendorfer R, Ruckstuhl N, D’Amato M, 500. Mietlicki-Baase EG, Ortinski PI, Rupprecht LE, Olivos DR, Alhadeff AL, Pierce RC,
Rovati L, Beglinger C. The role of long chain fatty acids in regulating food intake and Hayes MR. The food intake-suppressive effects of glucagon-like peptide-1 receptor
cholecystokinin release in humans. Gut 46: 688 – 693, 2000. signaling in the ventral tegmental area are mediated by AMPA/kainate receptors. Am
J Physiol Endocrinol Metab 305: E1367–E1374, 2013.
480. Matzinger D, Gutzwiller JP, Drewe J, Orban A, Engel R, D’Amato M, Rovati L, Be-
glinger C. Inhibition of food intake in response to intestinal lipid is mediated by 501. Miller LJ, Holicky EL, Ulrich CD, Wieben ED. Abnormal processing of the human
cholecystokinin in humans. Am J Physiol Regul Integr Comp Physiol 277: R1718 –R1724, cholecystokinin receptor gene in association with gallstones and obesity. Gastroenter-
1999. ology 109: 1375–1380, 1995.
502. Mirshahi UL, Still CD, Masker KK, Gerhard GS, Carey DJ, Mirshahi T. The 523. Muir JG, Lu ZX, Young GP, Cameron-Smith D, Collier GR, O’Dea K. Resistant starch
MC4R(I251L) allele is associated with better metabolic status and more weight loss in the diet increases breath hydrogen and serum acetate in human subjects. Am J Clin
after gastric bypass surgery. J Clin Endocrinol Metab 96: E2088 –2096, 2011. Nutr 61: 792–799, 1995.
503. Miyamoto Y, Miyamoto M. Immunohistochemical localizations of secretin, chole- 524. Muller TD, Nogueiras R, Andermann ML, Andrews ZB, Anker SD, Argente J, Batter-
cystokinin, and somatostatin in the rat small intestine after acute cisplatin treatment. ham RL, Benoit SC, Bowers CY, Broglio F, Casanueva FF, D’Alessio D, Depoortere I,
Exp Mol Pathol 77: 238 –245, 2004. Geliebter A, Ghigo E, Cole PA, Cowley M, Cummings DE, Dagher A, Diano S,
Dickson SL, Dieguez C, Granata R, Grill HJ, Grove K, Habegger KM, Heppner K,
504. Miyasaka K, Guan DF, Liddle RA, Green GM. Feedback regulation by trypsin: evi- Heiman ML, Holsen L, Holst B, Inui A, Jansson JO, Kirchner H, Korbonits M, Laferrere
dence for intraluminal CCK-releasing peptide. Am J Physiol Gastrointest Liver Physiol B, LeRoux CW, Lopez M, Morin S, Nakazato M, Nass R, Perez-Tilve D, Pfluger PT,
257: G175–G181, 1989.
Schwartz TW, Seeley RJ, Sleeman M, Sun Y, Sussel L, Tong J, Thorner MO, van der
505. Modigliani R, Bernier JJ. Absorption of glucose, sodium, and water by the human Lely AJ, van der Ploeg LH, Zigman JM, Kojima M, Kangawa K, Smith RG, Horvath T,
jejunum studied by intestinal perfusion with a proximal occluding balloon and at Tschop MH. Ghrelin. Mol Metab 4: 437– 460, 2015.
variable flow rates. Gut 12: 184 –193, 1971.
525. Mumphrey MB, Patterson LM, Zheng H, Berthoud HR. Roux-en-Y gastric bypass
506. Mohlig M, Spranger J, Otto B, Ristow M, Tschop M, Pfeiffer AF. Euglycemic hyperin- surgery increases number but not density of CCK-, GLP-1-, 5-HT-, and neuro-
sulinemia, but not lipid infusion, decreases circulating ghrelin levels in humans. J En- tensin-expressing enteroendocrine cells in rats. Neurogastroenterol Motil 25: e70 –
docrinol Invest 25: RC36 –38, 2002. 79, 2013.
507. Mokadem M, Zechner JF, Margolskee RF, Drucker DJ, Aguirre V. Effects of Roux- 526. Murdolo G, Lucidi P, Di Loreto C, Parlanti N, De Cicco A, Fatone C, Fanelli CG, Bolli
en-Y gastric bypass on energy and glucose homeostasis are preserved in two mouse GB, Santeusanio F, De Feo P. Insulin is required for prandial ghrelin suppression in
models of functional glucagon-like peptide-1 deficiency. Mol Metab 3: 191–201, 2014. humans. Diabetes 52: 2923–2927, 2003.
508. Moller JB, Jusko WJ, Gao W, Hansen T, Pedersen O, Holst JJ, Overgaard RV, Madsen 527. Mussig K, Staiger H, Machicao F, Haring HU, Fritsche A. Genetic variants affecting
H, Ingwersen SH. Mechanism-based population modelling for assessment of L-cell incretin sensitivity and incretin secretion. Diabetologia 53: 2289 –2297, 2010.
function based on total GLP-1 response following an oral glucose tolerance test. J
528. Muurahainen NE, Kissileff HR, Lachaussée J, Pi-Sunyer FX. Effect of a soup preload on
Pharmacokinet Pharmacodyn 38: 713–725, 2011.
reduction of food intake by cholecystokinin in humans. Am J Physiol Regul Integr Comp
509. Monnier L, Lapinski H, Colette C. Contributions of fasting and postprandial plasma Physiol 260: R672–R680, 1991.
glucose increments to the overall diurnal hyperglycemia of type 2 diabetic patients:
variations with increasing levels of HbA(1c). Diabetes Care 26: 881– 885, 2003. 529. Myronovych A, Kirby M, Ryan KK, Zhang W, Jha P, Setchell KD, Dexheimer PJ,
Aronow B, Seeley RJ, Kohli R. Vertical sleeve gastrectomy reduces hepatic steatosis
510. Monteleone P, Bencivenga R, Longobardi N, Serritella C, Maj M. Differential re- while increasing serum bile acids in a weight-loss-independent manner. Obesity 22:
sponses of circulating ghrelin to high-fat or high-carbohydrate meal in healthy women. 390 – 400, 2014.
J Clin Endocrinol Metab 88: 5510 –5514, 2003.
530. Nagaya N, Kojima M, Uematsu M, Yamagishi M, Hosoda H, Oya H, Hayashi Y,
511. Monteleone P, Serritella C, Martiadis V, Maj M. Deranged secretion of ghrelin and Kangawa K. Hemodynamic and hormonal effects of human ghrelin in healthy volun-
obestatin in the cephalic phase of vagal stimulation in women with anorexia nervosa. teers. Am J Physiol Regul Integr Comp Physiol 280: R1483–R1487, 2001.
Biol Psychiatry 64: 1005–1008, 2008.
531. Nagell CF, Pedersen JF, Holst JJ. The antagonistic metabolite of GLP-1, GLP-1 (9 –
512. Monteleone P, Serritella C, Scognamiglio P, Maj M. Enhanced ghrelin secretion in the 36)amide, does not influence gastric emptying and hunger sensations in man. Scand J
cephalic phase of food ingestion in women with bulimia nervosa. Psychoneuroendocri- Gastroenterol 42: 28 –33, 2007.
nology 35: 284 –288, 2010.
532. Nair NS, Brennan IM, Little TJ, Gentilcore D, Hausken T, Jones KL, Wishart JM,
513. Moran TH, Dailey MJ. Intestinal feedback signaling and satiety. Physiol Behav 105: Horowitz M, Feinle-Bisset C. Reproducibility of energy intake, gastric emptying,
77– 81, 2011. blood glucose, plasma insulin and cholecystokinin responses in healthy young males.
Br J Nutr 101: 1094 –1102, 2009.
514. Moran TH, Katz LF, Plata-Salaman CR, Schwartz GJ. Disordered food intake and
obesity in rats lacking cholecystokinin A receptors. Am J Physiol Regul Integr Comp 533. Nannipieri M, Baldi S, Mari A, Colligiani D, Guarino D, Camastra S, Barsotti E, Berta
Physiol 274: R618 –R625, 1998. R, Moriconi D, Bellini R, Anselmino M, Ferrannini E. Roux-en-Y gastric bypass and
sleeve gastrectomy: mechanisms of diabetes remission and role of gut hormones. J
515. Moran TH, Robinson PH, Goldrich MS, McHugh PR. Two brain cholecystokinin re-
Clin Endocrinol Metab 98: 4391– 4399, 2013.
ceptors: implications for behavioral actions. Brain Res 362: 175–179, 1986.
534. Naslund E, Barkeling B, King N, Gutniak M, Blundell JE, Holst JJ, Rossner S, Hellstrom
516. Moran TH, Shnayder L, Hostetler AM, McHugh PR. Pylorectomy reduces the satiety
PM. Energy intake and appetite are suppressed by glucagon-like peptide-1 (GLP-1) in
action of cholecystokinin. Am J Physiol Regul Integr Comp Physiol 255: R1059 –R1063,
obese men. Int J Obes Relat Metab Disord 23: 304 –311, 1999.
1988.
535. Näslund E, Gutniak M, Skogar S, Rössner S, Hellström PM. Glucagon-like peptide 1
517. Moran TH, Smedh U, Kinzig KP, Scott KA, Knipp S, Ladenheim EE. Peptide YY(3–36)
increases the period of postprandial satiety and slows gastric emptying in obese men.
inhibits gastric emptying and produces acute reductions in food intake in rhesus
Am J Clin Nutr 68: 525–530, 1998.
monkeys. Am J Physiol Regul Integr Comp Physiol 288: R384 –R388, 2005.
536. Naslund I, Beckman KW. Gastric emptying rate after gastric bypass and gastroplasty.
518. Morinigo R, Moize V, Musri M, Lacy AM, Navarro S, Marin JL, Delgado S, Casamitjana
Scand J Gastroenterol 22: 193–201, 1987.
R, Vidal J. Glucagon-like peptide-1, peptide YY, hunger, and satiety after gastric bypass
surgery in morbidly obese subjects. J Clin Endocrinol Metab 91: 1735–1740, 2006. 537. Nauck MA, Bartels E, Orskov C, Ebert R, Creutzfeldt W. Additive insulinotropic
effects of exogenous synthetic human gastric inhibitory polypeptide and glucagon-like
519. Moriya R, Shirakura T, Ito J, Mashiko S, Seo T. Activation of sodium-glucose cotrans-
porter 1 ameliorates hyperglycemia by mediating incretin secretion in mice. Am J peptide-1-(7–36) amide infused at near-physiological insulinotropic hormone and
Physiol Endocrinol Metab 297: E1358 –E1365, 2009. glucose concentrations. J Clin Endocrinol Metab 76: 912–917, 1993.
520. Mortensen K, Christensen LL, Holst JJ, Orskov C. GLP-1 and GIP are colocalized in a 538. Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W. Preserved
subset of endocrine cells in the small intestine. Regul Pept 114: 189 –196, 2003. incretin activity of glucagon-like peptide 1 [7–36 amide] but not of synthetic human
gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 91:
521. Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW. Central nervous 301–307, 1993.
system control of food intake and body weight. Nature 443: 289 –295, 2006.
539. Nauck MA, Homberger E, Siegel EG, Allen RC, Eaton RP, Ebert R, Creutzfeldt W.
522. Moss C, Dhillo WS, Frost G, Hickson M. Gastrointestinal hormones: the regulation of Incretin effects of increasing glucose loads in man calculated from venous insulin and
appetite and the anorexia of ageing. J Hum Nutr Diet 25: 3–15, 2012. C-peptide responses. J Clin Endocrinol Metab 63: 492– 498, 1986.
540. Nauck MA, Kemmeries G, Holst JJ, Meier JJ. Rapid tachyphylaxis of the glucagon-like 559. Padmanabhan P, Kumar A, Kumar S, Chaudhary RK, Gulyas B. Nanoparticles in
peptide 1-induced deceleration of gastric emptying in humans. Diabetes 60: 1561– practice for molecular-imaging applications: an overview. Acta Biomater 41: 1–16,
1565, 2011. 2016.
541. Nauck MA, Niedereichholz U, Ettler R, Holst JJ, Orskov C, Ritzel R, Schmiegel WH. 560. Page AJ, Symonds E, Peiris M, Blackshaw LA, Young RL. Peripheral neural targets in
Glucagon-like peptide 1 inhibition of gastric emptying outweighs its insulinotropic obesity. Br J Pharmacol 166: 1537–1558, 2012.
effects in healthy humans. Am J Physiol Endocrinol Metab 273: E981–E988, 1997.
561. Panaro BL, Tough IR, Engelstoft MS, Matthews RT, Digby GJ, Moller CL, Svendsen B,
542. Nauck MA, Vardarli I, Deacon CF, Holst JJ, Meier JJ. Secretion of glucagon-like pep- Gribble F, Reimann F, Holst JJ, Holst B, Schwartz TW, Cox HM, Cone RD. The
tide-1 (GLP-1) in type 2 diabetes: what is up, what is down? Diabetologia 54: 10 –18, melanocortin-4 receptor is expressed in enteroendocrine L cells and regulates the
2011. release of peptide YY and glucagon-like peptide 1 in vivo. Cell Metab 20: 1018 –1029,
2014.
543. Nelson G, Chandrashekar J, Hoon MA, Feng L, Zhao G, Ryba NJ, Zuker CS. An
562. Park MI, Camilleri M, O’Connor H, Oenning L, Burton D, Stephens D, Zinsmeister
amino-acid taste receptor. Nature 416: 199 –202, 2002.
AR. Effect of different macronutrients in excess on gastric sensory and motor func-
544. Nguyen NQ, Debreceni TL, Bambrick JE, Bellon M, Wishart J, Standfield S, Rayner tions and appetite in normal-weight, overweight, and obese humans. Am J Clin Nutr
CK, Horowitz M. Rapid gastric and intestinal transit is a major determinant of changes 85: 411– 418, 2007.
in blood glucose, intestinal hormones, glucose absorption, and postprandial symp- 563. Parker BA, Doran S, Wishart J, Horowitz M, Chapman IM. Effects of small intestinal
toms after gastric bypass. Obesity 22: 2003–2009, 2014. and gastric glucose administration on the suppression of plasma ghrelin concentra-
tions in healthy older men and women. Clin Endocrinol 62: 539 –546, 2005.
545. Nguyen NQ, Debreceni TL, Burgstad CM, Neo M, Bellon M, Wishart JM, Standfield
S, Bartholomeusz D, Rayner CK, Wittert G, Horowitz M. Effects of fat and protein 564. Parker HE, Wallis K, le Roux CW, Wong KY, Reimann F, Gribble FM. Molecular
preloads on pouch emptying, intestinal transit, glycaemia, gut hormones, glucose mechanisms underlying bile acid-stimulated glucagon-like peptide-1 secretion. Br J
absorption, blood pressure and gastrointestinal symptoms after Roux-en-Y gastric Pharmacol 165: 414 – 423, 2012.
bypass. Obes Surg 26: 77– 84, 2016.
565. Patterson LM, Zheng H, Ward SM, Berthoud HR. Immunohistochemical identification
546. Nicolaus M, Brödl J, Linke R, Woerle HJ, Göke B, Schirra J. Endogenous GLP-1 of cholecystokinin A receptors on interstitial cells of Cajal, smooth muscle, and enteric
regulates postprandial glycemia in humans: relative contributions of insulin, glucagon, neurons in rat pylorus. Cell Tissue Res 305: 11–23, 2001.
and gastric emptying. J Clin Endocrinol Metab 96: 229 –236, 2011.
566. Patti ME, Houten SM, Bianco AC, Bernier R, Larsen PR, Holst JJ, Badman MK, Mara-
547. Niederau C, Schwarzendrube J, Luthen R, Niederau M, Strohmeyer G, Rovati L. tos-Flier E, Mun EC, Pihlajamaki J, Auwerx J, Goldfine AB. Serum bile acids are higher
Effects of cholecystokinin receptor blockade on circulating concentrations of glucose, in humans with prior gastric bypass: potential contribution to improved glucose and
insulin, C-peptide, and pancreatic polypeptide after various meals in healthy human lipid metabolism. Obesity 17: 1671–1677, 2009.
volunteers. Pancreas 7: 1–10, 1992.
566a.Pavlov IP. The Work of the Digestive Glands, translated by Thompson WH. London,
548. Nishizawa M, Moore MC, Shiota M, Gustavson SM, Snead WL, Neal DW, Cher- UK: Griffin, 1910.
rington AD. Effect of intraportal glucagon-like peptide-1 on glucose metabolism in
567. Pearse AG, Coulling I, Weavers B, Friesen S. The endocrine polypeptide cells of the
conscious dogs. Am J Physiol Endocrinol Metab 284: E1027–E1036, 2003. human stomach, duodenum, and jejunum. Gut 11: 649 – 658, 1970.
549. Nolan LJ, Guss JL, Liddle RA, Pi-Sunyer FX, Kissileff HR. Elevated plasma cholecysto- 568. Pedersen J, Ugleholdt RK, Jorgensen SM, Windelov JA, Grunddal KV, Schwartz TW,
kinin and appetitive ratings after consumption of a liquid meal in humans. Nutrition 19: Fuchtbauer EM, Poulsen SS, Holst PJ, Holst JJ. Glucose metabolism is altered after loss
553–557, 2003. of L cells and alpha-cells but not influenced by loss of K cells. Am J Physiol Endocrinol
Metab 304: E60 –E73, 2013.
550. O’Donovan DG, Doran S, Feinle-Bisset C, Jones KL, Meyer JH, Wishart JM, Morris
HA, Horowitz M. Effect of variations in small intestinal glucose delivery on plasma 569. Penagini R, Misiewicz JJ, Frost PG. Effect of jejunal infusion of bile acids on small bowel
glucose, insulin, and incretin hormones in healthy subjects and type 2 diabetes. J Clin transit and fasting jejunal motility in man. Gut 29: 789 –794, 1988.
Endocrinol Metab 89: 3431–3435, 2004.
570. Penney NC, Kinross J, Newton RC, Purkayastha S. The role of bile acids in reducing
551. Odstrcil EA, Martinez JG, Santa Ana CA, Xue B, Schneider RE, Steffer KJ, Porter JL, the metabolic complications of obesity after bariatric surgery: a systematic review. Int
Asplin J, Kuhn JA, Fordtran JS. The contribution of malabsorption to the reduction in J Obes (Lond) 39: 1565–1574, 2015.
net energy absorption after long-limb Roux-en-Y gastric bypass. Am J Clin Nutr 92:
704 –713, 2010. 571. Perello M, Dickson SL. Ghrelin signalling on food reward: a salient link between the
gut and the mesolimbic system. J Neuroendocrinol 27: 424 – 434, 2015.
552. Oesch S, Degen L, Beglinger C. Effect of a protein preload on food intake and satiety
572. Perez-Tilve D, Hofmann SM, Basford J, Nogueiras R, Pfluger PT, Patterson JT, Grant
feelings in response to duodenal fat perfusions in healthy male subjects. Am J Physiol
E, Wilson-Perez HE, Granholm NA, Arnold M, Trevaskis JL, Butler AA, Davidson WS,
Regul Integr Comp Physiol 289: R1042–R1047, 2005.
Woods SC, Benoit SC, Sleeman MW, DiMarchi RD, Hui DY, Tschop MH. Melano-
553. Ohlsson L, Kohan AB, Tso P, Ahren B. GLP-1 released to the mesenteric lymph duct cortin signaling in the CNS directly regulates circulating cholesterol. Nat Neurosci 13:
in mice: effects of glucose and fat. Regul Pept 189: 40 – 45, 2014. 877– 882, 2010.
573. Persaud SJ, Bewick GA. Peptide YY: more than just an appetite regulator. Diabetologia
554. Ohno T, Kamiyama Y, Aihara R, Nakabayashi T, Mochiki E, Asao T, Kuwano H.
57: 1762–1769, 2014.
Ghrelin does not stimulate gastrointestinal motility and gastric emptying: an experi-
mental study of conscious dogs. Neurogastroenterol Motil 18: 129 –135, 2006. 574. Peterli R, Steinert RE, Woelnerhanssen B, Peters T, Christoffel-Courtin C, Gass M,
Kern B, von Fluee M, Beglinger C. Metabolic and hormonal changes after laparoscopic
555. Okano-Matsumoto S, McRoberts JA, Tache Y, Adelson DW. Electrophysiological
Roux-en-Y gastric bypass and sleeve gastrectomy: a randomized, prospective trial.
evidence for distinct vagal pathways mediating CCK-evoked motor effects in the Obes Surg 22: 740 –748, 2012.
proximal versus distal stomach. J Physiol 589: 371–393, 2011.
575. Pfeiffer A, Schmidt T, Vidon N, Kaess H. Effect of ethanol on absorption of a nutrient
556. Oliver G, Schafer EA. The physiological effects of extracts of the suprarenal capsules. solution in the upper human intestine. Scand J Gastroenterol 28: 515–521, 1993.
J Physiol 18: 230 –276, 1895.
576. Pfluger PT, Kampe J, Castaneda TR, Vahl T, D’Alessio DA, Kruthaupt T, Benoit SC,
557. Orskov C, Wettergren A, Holst JJ. Secretion of the incretin hormones glucagon-like Cuntz U, Rochlitz HJ, Moehlig M, Pfeiffer AF, Koebnick C, Weickert MO, Otto B,
peptide-1 and gastric inhibitory polypeptide correlates with insulin secretion in nor- Spranger J, Tschop MH. Effect of human body weight changes on circulating levels of
mal man throughout the day. Scand J Gastroenterol 31: 665– 670, 1996. peptide YY and peptide YY3-36. J Clin Endocrinol Metab 92: 583–588, 2007.
558. Overduin J, Frayo RS, Grill HJ, Kaplan JM, Cummings DE. Role of the duodenum and 577. Phillips LK, Deane AM, Jones KL, Rayner CK, Horowitz M. Gastric emptying and
macronutrient type in ghrelin regulation. Endocrinology 146: 845– 850, 2005. glycaemia in health and diabetes mellitus. Nat Rev Endocrinol 11: 112–128, 2015.
578. Phillips LK, Deane AM, Jones KL, Rayner CK, Horowitz M. Gastric emptying and 598. Punjabi M, Arnold M, Geary N, Langhans W, Pacheco-Lopez G. Peripheral glucagon-
glycaemia in health and diabetes mellitus. Nat Rev Endocrinol 11: 112–128, 2015. like peptide-1 (GLP-1) and satiation. Physiol Behav 105: 71–76, 2011.
579. Phillips RJ, Powley TL. Tension and stretch receptors in gastrointestinal smooth mus- 599. Punjabi M, Arnold M, Ruttimann E, Graber M, Geary N, Pacheco-Lopez G, Langhans
cle: re-evaluating vagal mechanoreceptor electrophysiology. Brain Res 34: 1–26, 2000. W. Circulating glucagon-like peptide-1 (GLP-1) inhibits eating in male rats by acting in
the hindbrain and without inducing avoidance. Endocrinology 155: 1690 –1699, 2014.
580. Phillips WT, Schwartz JG, McMahan CA. Reduced postprandial blood glucose levels in
recently diagnosed non-insulin-dependent diabetics secondary to pharmacologically 600. Raben A, Agerholm-Larsen L, Flint A, Holst JJ, Astrup A. Meals with similar energy
induced delayed gastric emptying. Dig Dis Sci 38: 51–58, 1993. densities but rich in protein, fat, carbohydrate, or alcohol have different effects on
energy expenditure and substrate metabolism but not on appetite and energy intake.
581. Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M, Lau DC, le Roux
Am J Clin Nutr 77: 91–100, 2003.
CW, Violante Ortiz R, Jensen CB, Wilding JP, Obesity S, Prediabetes NNSG. A ran-
domized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med 601. Radziuk J, Kemmer F, Morishima T, Berchtold P, Vranic M. The effects of an alpha-
373: 11–22, 2015. glucoside hydrolase inhibitor on glycemia and the absorption of sucrose in man de-
termined using a tracer method. Diabetes 33: 207–213, 1984.
582. Pierson ME, Comstock JM, Simmons RD, Kaiser F, Julien R, Zongrone J, Rosamond
JD. Synthesis and biological evaluation of potent, selective, hexapeptide CCK-A ago- 602. Ramsay DS, Woods SC. Clarifying the roles of homeostasis and allostasis in physio-
nist anorectic agents. J Med Chem 40: 4302– 4307, 1997. logical regulation. Psychol Rev 121: 225–247, 2014.
583. Pilichiewicz AN, Chaikomin R, Brennan IM, Wishart JM, Rayner CK, Jones KL, Smout 603. Ranganath LR, Beety JM, Morgan LM, Wright JW, Howland R, Marks V. Attenuated
AJPM, Horowitz M, Feinle-Bisset C. Load-dependent effects of duodenal glucose on GLP-1 secretion in obesity: cause or consequence? Gut 38: 916 –919, 1996.
glycemia, gastrointestinal hormones, antropyloroduodenal motility, and energy intake
in healthy men. Am J Physiol Endocrinol Metab 293: E743–E753, 2007. 604. Rask E, Olsson T, Soderberg S, Johnson O, Seckl J, Holst JJ, Ahren B, Northern
Sweden Monitoring of Trends and Determinants in Cardiovascular Disease. Impaired
584. Pilichiewicz AN, Papadopoulos P, Brennan IM, Little TJ, Meyer JH, Wishart JM, incretin response after a mixed meal is associated with insulin resistance in nondia-
Horowitz M, Feinle-Bisset C. Load-dependent effects of duodenal lipid on antropy-
betic men. Diabetes Care 24: 1640 –1645, 2001.
loroduodenal motility, plasma CCK and PYY, and energy intake in healthy men. Am J
Physiol Regul Integr Comp Physiol 293: R2170 –R2178, 2007. 605. Rasmussen BA, Breen DM, Luo P, Cheung GW, Yang CS, Sun B, Kokorovic A, Rong
W, Lam TK. Duodenal activation of cAMP-dependent protein kinase induces vagal
585. Pironi L, Stanghellini V, Miglioli M, Corinaldesi R, De Giorgio R, Ruggeri E, Tosetti C,
afferent firing and lowers glucose production in rats. Gastroenterology 142: 834 – 843,
Poggioli G, Morselli Labate AM, Monetti N, et al. Fat-induced ileal brake in humans: a
2012.
dose-dependent phenomenon correlated to the plasma levels of peptide YY. Gastro-
enterology 105: 733–739, 1993. 606. Rayner CK, Horowitz M. Physiology of the ageing gut. Curr Opin Clin Nutr Metab Care
16: 33–38, 2013.
586. Plaisancie P, Bernard C, Chayvialle JA, Cuber JC. Release of peptide YY by neu-
rotransmitters and gut hormones in the isolated, vascularly perfused rat colon. Scand 607. Reeve JR Jr, Eysselein VE, Rosenquist G, Zeeh J, Regner U, Ho FJ, Chew P, Davis MT,
J Gastroenterol 30: 568 –574, 1995. Lee TD, Shively JE, Brazer SR, Liddle RA. Evidence that CCK-58 has structure that
influences its biological activity. Am J Physiol Gastrointest Liver Physiol 270: G860 –
587. Plamboeck A, Veedfald S, Deacon CF, Hartmann B, Wettergren A, Svendsen LB,
Meisner S, Hovendal C, Knop FK, Vilsboll T, Holst JJ. Characterisation of oral and iv G868, 1996.
glucose handling in truncally vagotomised subjects with pyloroplasty. Eur J Endocrinol
608. Reeve JR Jr, Wu SV, Keire DA, Faull K, Chew P, Solomon TE, Green GM, Coskun T.
169: 187–201, 2013.
Differential bile-pancreatic secretory effects of CCK-58 and CCK-8. Am J Physiol
588. Plamboeck A, Veedfald S, Deacon CF, Hartmann B, Wettergren A, Svendsen LB, Gastrointest Liver Physiol 286: G395–G402, 2004.
Meisner S, Hovendal C, Vilsboll T, Knop FK, Holst JJ. The effect of exogenous GLP-1
609. Rehfeld JF. Accurate measurement of cholecystokinin in plasma. Clin Chem 44: 991–
on food intake is lost in male truncally vagotomized subjects with pyloroplasty. Am J
1001, 1998.
Physiol Gastrointest Liver Physiol 304: G1117–G1127, 2013.
610. Rehfeld JF. Beginnings: a reflection on the history of gastrointestinal endocrinology.
589. Polak JM, Bloom S, Coulling I, Pearse AG. Immunofluorescent localization of entero-
Regul Pept 177 Suppl: S1–5, 2012.
glucagon cells in the gastrointestinal tract of the dog. Gut 12: 311–318, 1971.
611. Rehfeld JF. A centenary of gastrointestinal endocrinology. Horm Metab Res 36: 735–
590. Polak JM, Bloom SR, Rayford PL, Pearse AG, Buchan AM, Thompson JC. Identification
741, 2004.
of cholecystokinin-secreting cells. Lancet 2: 1016 –1018, 1975.
612. Rehfeld JF, Friis-Hansen L, Goetze JP, Hansen TV. The biology of cholecystokinin and
591. Polak JM, Pearse AG, Heath CM. Complete identification of endocrine cells in the
gastrointestinal tract using semithin-thin sections to identify motilin cells in human and gastrin peptides. Curr Top Med Chem 7: 1154 –1165, 2007.
animal intestine. Gut 16: 225–229, 1975.
613. Reidelberger R, Haver A, Anders K, Apenteng B. Role of capsaicin-sensitive peripheral
592. Polonsky KS, Given BD, Hirsch L, Shapiro ET, Tillil H, Beebe C, Galloway JA, Frank sensory neurons in anorexic responses to intravenous infusions of cholecystokinin,
BH, Karrison T, Van Cauter E. Quantitative study of insulin secretion and clearance in peptide YY-(3–36), and glucagon-like peptide-1 in rats. Am J Physiol Endocrinol Metab
normal and obese subjects. J Clin Invest 81: 435– 441, 1988. 307: E619 –E629, 2014.
593. Polonsky KS, Given BD, Van Cauter E. Twenty-four-hour profiles and pulsatile pat- 614. Reidelberger R, Haver A, Chelikani PK. Role of peptide YY(3–36) in the satiety pro-
terns of insulin secretion in normal and obese subjects. J Clin Invest 81: 442– 448, 1988. duced by gastric delivery of macronutrients in rats. Am J Physiol Endocrinol Metab 304:
E944 –E950, 2013.
594. Prigeon RL, Quddusi S, Paty B, D’Alessio DA. Suppression of glucose production by
GLP-1 independent of islet hormones: a novel extrapancreatic effect. Am J Physiol 615. Reidelberger RD, Hernandez J, Fritzsch B, Hulce M. Abdominal vagal mediation of the
Endocrinol Metab 285: E701–E707, 2003. satiety effects of CCK in rats. Am J Physiol Regul Integr Comp Physiol 286: R1005–
R1012, 2004.
595. Pritchett CE, Hajnal A. Glucagon-like peptide-1 regulation of carbohydrate intake is
differentially affected by obesogenic diets. Obesity 20: 313–317, 2012. 616. Reidelberger RD, Varga G, Liehr RM, Castellanos DA, Rosenquist GL, Wong HC,
Walsh JH. Cholecystokinin suppresses food intake by a nonendocrine mechanism in
596. Prudom C, Liu J, Patrie J, Gaylinn BD, Foster-Schubert KE, Cummings DE, Thorner rats. Am J Physiol Regul Integr Comp Physiol 267: R901–R908, 1994.
MO, Geysen HM. Comparison of competitive radioimmunoassays and two-site sand-
wich assays for the measurement and interpretation of plasma ghrelin levels. J Clin 617. Reimann F. Molecular mechanisms underlying nutrient detection by incretin-secreting
Endocrinol Metab 95: 2351–2358, 2010. cells. Int Dairy J 20: 236 –242, 2010.
597. Psichas A, Reimann F, Gribble FM. Gut chemosensing mechanisms. J Clin Invest 125: 618. Reimann F, Habib AM, Tolhurst G, Parker HE, Rogers GJ, Gribble FM. Glucose
908 –917, 2015. sensing in L cells: a primary cell study. Cell Metab 8: 532–539, 2008.
619. Richard JE, Anderberg RH, Goteson A, Gribble FM, Reimann F, Skibicka KP. Activa- 640. Ryan AT, Feinle-Bisset C, Kallas A, Wishart JM, Clifton PM, Horowitz M, Luscombe-
tion of the GLP-1 receptors in the nucleus of the solitary tract reduces food reward Marsh ND. Intraduodenal protein modulates antropyloroduodenal motility, hormone
behavior and targets the mesolimbic system. PLoS One 10: e0119034, 2015. release, glycemia, appetite, and energy intake in lean men. Am J Clin Nutr 96: 474 –
482, 2012.
620. Richard JE, Farkas I, Anesten F, Anderberg RH, Dickson SL, Gribble FM, Reimann F,
Jansson JO, Liposits Z, Skibicka KP. GLP-1 receptor stimulation of the lateral para- 641. Ryan AT, Luscombe-Marsh ND, Saies AA, Little TJ, Standfield S, Horowitz M, Feinle-
brachial nucleus reduces food intake: neuroanatomical, electrophysiological, and be- Bisset C. Effects of intraduodenal lipid and protein on gut motility and hormone
havioral evidence. Endocrinology 155: 4356 – 4367, 2014. release, glycemia, appetite, and energy intake in lean men. Am J Clin Nutr 98: 300 –311,
2013.
621. Riddle M, Umpierrez G, DiGenio A, Zhou R, Rosenstock J. Contributions of basal and
postprandial hyperglycemia over a wide range of A1C levels before and after treat- 642. Ryan KK, Tremaroli V, Clemmensen C, Kovatcheva-Datchary P, Myronovych A,
ment intensification in type 2 diabetes. Diabetes Care 34: 2508 –2514, 2011. Karns R, Wilson-Perez HE, Sandoval DA, Kohli R, Backhed F, Seeley RJ. FXR is a
622. Rieu PN, Jansen JB, Hopman WP, Joosten HJ, Lamers CB. Effect of partial gastrectomy molecular target for the effects of vertical sleeve gastrectomy. Nature 509: 183–188,
with Billroth II or Roux-en-Y anastomosis on postprandial and cholecystokinin-stim- 2014.
ulated gallbladder contraction and secretion of cholecystokinin and pancreatic poly-
643. Saad A, Dalla Man C, Nandy DK, Levine JA, Bharucha AE, Rizza RA, Basu R, Carter RE,
peptide. Dig Dis Sci 35: 1066 –1072, 1990.
Cobelli C, Kudva YC, Basu A. Diurnal pattern to insulin secretion and insulin action in
623. Rinaman L. Ascending projections from the caudal visceral nucleus of the solitary tract healthy individuals. Diabetes 61: 2691–2700, 2012.
to brain regions involved in food intake and energy expenditure. Brain Res 1350:
644. Saad MF, Bernaba B, Hwu CM, Jinagouda S, Fahmi S, Kogosov E, Boyadjian R. Insulin
18 –34, 2010.
regulates plasma ghrelin concentration. J Clin Endocrinol Metab 87: 3997– 4000, 2002.
624. Rinaman L. Hindbrain contributions to anorexia. Am J Physiol Regul Integr Comp Physiol
287: R1035–R1036, 2004. 645. Salaun PY, Querellou S, Nguyen JM, Bodet-Milin C, Carlier T, Turzo A, Bizais Y,
Couturier O. Comparison of gastric emptying scintigraphy based on the geometric
625. Rinaman L. Hindbrain noradrenergic A2 neurons: diverse roles in autonomic, endo- mean of the gastric proportion of the abdominal radioactivity or on the geometric
crine, cognitive, and behavioral functions. Am J Physiol Regul Integr Comp Physiol 300: mean of the intragastric radioactivity. Nucl Med Commun 27: 431– 437, 2006.
R222–R235, 2011.
646. Salehi M, D’Alessio DA. Effects of glucagon like peptide-1 to mediate glycemic effects
626. Roberts K, Duffy A, Kaufman J, Burrell M, Dziura J, Bell R. Size matters: gastric pouch of weight loss surgery. Rev Endocr Metab Disord 15: 171–179, 2014.
size correlates with weight loss after laparoscopic Roux-en-Y gastric bypass. Surg
Endosc 21: 1397–1402, 2007. 647. Salehi M, Gastaldelli A, D’Alessio DA. Altered islet function and insulin clearance cause
hyperinsulinemia in gastric bypass patients with symptoms of postprandial hypogly-
627. Roberts RE, Glicksman C, Alaghband-Zadeh J, Sherwood RA, Akuji N, le Roux CW. cemia. J Clin Endocrinol Metab 99: 2008 –2017, 2014.
The relationship between postprandial bile acid concentration, GLP-1, PYY and ghre-
lin. Clin Endocrinol 74: 67–72, 2011. 648. Salehi M, Gastaldelli A, D’Alessio DA. Blockade of glucagon-like peptide 1 receptor
corrects postprandial hypoglycemia after gastric bypass. Gastroenterology 146: 669 –
628. Rocca AS, Brubaker PL. Role of the vagus nerve in mediating proximal nutrient- 680, 2014.
induced glucagon-like peptide-1 secretion. Endocrinology 140: 1687–1694, 1999.
649. Salehi M, Prigeon RL, D’Alessio DA. Gastric bypass surgery enhances glucagon-like
629. Rolls BJ. What is the role of portion control in weight management? Int J Obes 38 Suppl
peptide 1-stimulated postprandial insulin secretion in humans. Diabetes 60: 2308 –
1: S1– 8, 2014.
2314, 2011.
630. Rolls BJ, Bell EA, Thorwart ML. Water incorporated into a food but not served with a
650. Salehi M, Vahl TP, D’Alessio DA. Regulation of islet hormone release and gastric
food decreases energy intake in lean women. Am J Clin Nutr 70: 448 – 455, 1999.
emptying by endogenous glucagon-like peptide 1 after glucose ingestion. J Clin Endo-
631. Rolls BJ, Bell EA, Waugh BA. Increasing the volume of a food by incorporating air crinol Metab 93: 4909 – 4916, 2008.
affects satiety in men. Am J Clin Nutr 72: 361–368, 2000.
651. Sam AH, Gunner DJ, King A, Persaud SJ, Brooks L, Hostomska K, Ford HE, Liu B,
632. Rolls BJ, Roe LS. Effect of the volume of liquid food infused intragastrically on satiety Ghatei MA, Bloom SR, Bewick GA. Selective ablation of peptide YY cells in adult mice
in women. Physiol Behav 76: 623– 631, 2002. reveals their role in beta cell survival. Gastroenterology 143: 459 – 468, 2012.
633. Roth CL, Enriori PJ, Harz K, Woelfle J, Cowley MA, Reinehr T. Peptide YY is a 652. Sandoval DA, Bagnol D, Woods SC, D’Alessio DA, Seeley RJ. Arcuate glucagon-like
regulator of energy homeostasis in obese children before and after weight loss. J Clin peptide 1 receptors regulate glucose homeostasis but not food intake. Diabetes 57:
Endocrinol Metab 90: 6386 – 6391, 2005. 2046 –2054, 2008.
634. Rubino F, Gagner M, Gentileschi P, Kini S, Fukuyama S, Feng J, Diamond E. The early 653. Sandoval DA, D’Alessio DA. Physiology of proglucagon peptides: role of glucagon and
effect of the Roux-en-Y gastric bypass on hormones involved in body weight regula- GLP-1 in health and disease. Physiol Rev 95: 513–548, 2015.
tion and glucose metabolism. Ann Surg 240: 236 –242, 2004.
654. Sanmiguel CP, Conklin JL, Cunneen SA, Barnett P, Phillips EH, Kipnes M, Pilcher J,
635. Runchey SS, Valsta LM, Schwarz Y, Wang C, Song X, Lampe JW, Neuhouser ML. Soffer EE. Gastric electrical stimulation with the TANTALUS System in obese type 2
Effect of low- and high-glycemic load on circulating incretins in a randomized clinical diabetes patients: effect on weight and glycemic control. J Diabetes Sci Technol 3:
trial. Metabolism 62: 188 –195, 2013. 964 –970, 2009.
636. Rushakoff RA, Goldfine ID, Beccaria LJ, Mathur A, Brand RJ, Liddle RA. Reduced 655. Santangelo A, Peracchi M, Conte D, Fraquelli M, Porrini M. Physical state of meal
postprandial cholecystokinin (CCK) secretion in patients with noninsulin-dependent affects gastric emptying, cholecystokinin release and satiety. Br J Nutr 80: 521–527,
diabetes mellitus: evidence for a role for CCK in regulating postprandial hyperglyce- 1998.
mia. J Clin Endocrinol Metab 76: 489 – 493, 1993.
656. Sarr MG, Billington CJ, Brancatisano R, Brancatisano A, Toouli J, Kow L, Nguyen NT,
637. Rushakoff RJ, Goldfine ID, Carter JD, Liddle RA. Physiological concentrations of cho-
Blackstone R, Maher JW, Shikora S, Reeds DN, Eagon JC, Wolfe BM, O’Rourke RW,
lecystokinin stimulate amino acid-induced insulin release in humans. J Clin Endocrinol
Fujioka K, Takata M, Swain JM, Morton JM, Ikramuddin S, Schweitzer M, Chand B,
Metab 65: 395– 401, 1987.
Rosenthal R, Group ES. The EMPOWER study: randomized, prospective, double-
638. Ruttimann EB, Arnold M, Geary N, Langhans W. GLP-1 antagonism with exendin blind, multicenter trial of vagal blockade to induce weight loss in morbid obesity. Obes
(9 –39) fails to increase spontaneous meal size in rats. Physiol Behav 100: 291–296, Surg 22: 1771–1782, 2012.
2010.
657. Sathananthan A, Man CD, Micheletto F, Zinsmeister AR, Camilleri M, Giesler PD,
639. Ruttimann EB, Arnold M, Hillebrand JJ, Geary N, Langhans W. Intrameal hepatic Laugen JM, Toffolo G, Rizza RA, Cobelli C, Vella A. Common genetic variation in
portal and intraperitoneal infusions of glucagon-like peptide-1 reduce spontaneous GLP1R and insulin secretion in response to exogenous GLP-1 in nondiabetic subjects:
meal size in the rat via different mechanisms. Endocrinology 150: 1174 –1181, 2009. a pilot study. Diabetes Care 33: 2074 –2076, 2010.
658. Sathananthan M, Ikramuddin S, Swain JM, Shah M, Piccinini F, Dalla Man C, Cobelli C, 677. Secher A, Jelsing J, Baquero AF, Hecksher-Sorensen J, Cowley MA, Dalboge LS,
Rizza RA, Camilleri M, Vella A. The effect of vagal nerve blockade using electrical Hansen G, Grove KL, Pyke C, Raun K, Schaffer L, Tang-Christensen M, Verma S,
impulses on glucose metabolism in nondiabetic subjects. Diabetes Metab Syndr Obes 7: Witgen BM, Vrang N, Bjerre Knudsen L. The arcuate nucleus mediates GLP-1 recep-
305–312, 2014. tor agonist liraglutide-dependent weight loss. J Clin Invest 124: 4473– 4488, 2014.
659. Savage AP, Adrian TE, Carolan G, Chatterjee VK, Bloom SR. Effects of peptide YY 678. Seeley RJ, Chambers AP, Sandoval DA. The role of gut adaptation in the potent effects
(PYY) on mouth to caecum intestinal transit time and on the rate of gastric emptying of multiple bariatric surgeries on obesity and diabetes. Cell Metab 21: 369 –378, 2015.
in healthy volunteers. Gut 28: 166 –170, 1987.
679. Seghieri M, Rebelos E, Gastaldelli A, Astiarraga BD, Casolaro A, Barsotti E, Pocai A,
660. Scheer FA, Morris CJ, Shea SA. The internal circadian clock increases hunger and Nauck M, Muscelli E, Ferrannini E. Direct effect of GLP-1 infusion on endogenous
appetite in the evening independent of food intake and other behaviors. Obesity 21: glucose production in humans. Diabetologia 56: 156 –161, 2013.
421– 423, 2013.
680. Seifarth C, Bergmann J, Holst JJ, Ritzel R, Schmiegel W, Nauck MA. Prolonged and
661. Schellekens H, De Francesco PN, Kandil D, Theeuwes WF, McCarthy T, van Oeffelen enhanced secretion of glucagon-like peptide 1 (7–36 amide) after oral sucrose due to
WE, Perello M, Giblin L, Dinan TG, Cryan JF. Ghrelin’s orexigenic effect is modulated alpha-glucosidase inhibition (acarbose) in Type 2 diabetic patients. Diabet Med 15:
via a serotonin 2C receptor interaction. ACS Chem Neurosci 6: 1186 –1197, 2015. 485– 491, 1998.
662. Schirra J, Houck P, Wank U, Arnold R, Goke B, Katschinski M. Effects of glucagon-like 681. Seimon RV, Brennan IM, Russo A, Little TJ, Jones KL, Standfield S, Wishart JM, Horow-
peptide-1(7–36)amide on antro-pyloro-duodenal motility in the interdigestive state itz M, Feinle-Bisset C. Gastric emptying, mouth-to-cecum transit, and glycemic, insu-
and with duodenal lipid perfusion in humans. Gut 46: 622– 631, 2000. lin, incretin, and energy intake responses to a mixed-nutrient liquid in lean, over-
weight, and obese males. Am J Physiol Endocrinol Metab 304: E294 –E300, 2013.
663. Schirra J, Katschinski M, Weidmann C, Schafer T, Wank U, Arnold R, Goke B. Gastric
emptying and release of incretin hormones after glucose ingestion in humans. J Clin 682. Seimon RV, Feltrin KL, Meyer JH, Brennan IM, Wishart JM, Horowitz M, Feinle-Bisset
Invest 97: 92–103, 1996. C. Effects of varying combinations of intraduodenal lipid and carbohydrate on antro-
pyloroduodenal motility, hormone release, and appetite in healthy males. Am J Physiol
664. Schirra J, Nicolaus M, Roggel R, Katschinski M, Storr M, Woerle HJ, Goke B. Endog- Regul Integr Comp Physiol 296: R912–R920, 2009.
enous glucagon-like peptide 1 controls endocrine pancreatic secretion and antro-
pyloro-duodenal motility in humans. Gut 55: 243–251, 2006. 683. Seimon RV, Lange K, Little TJ, Brennan IM, Pilichiewicz AN, Feltrin KL, Smeets AJ,
Horowitz M, Feinle-Bisset C. Pooled-data analysis identifies pyloric pressures and
665. Schirra J, Nicolaus M, Woerle HJ, Struckmeier C, Katschinski M, Goke B. GLP-1 plasma cholecystokinin concentrations as major determinants of acute energy intake
regulates gastroduodenal motility involving cholinergic pathways. Neurogastroenterol in healthy, lean men. Am J Clin Nutr 92: 61– 68, 2010.
Motil 21: 609 – 618, 2009.
684. Shah M, Law JH, Micheletto F, Sathananthan M, Man CD, Cobelli C, Rizza RA, Camil-
666. Schirra J, Wank U, Arnold R, Goke B, Katschinski M. Effects of glucagon-like peptide- leri M, Zinsmeister AR, Vella A. The contribution of endogenous glucagon-like pep-
1(7–36)amide on motility and sensation of the proximal stomach in humans. Gut 50: tide-1 to glucose metabolism after Roux-en-Y gastric bypass. Diabetes 63: 483– 493,
341–348, 2002. 2014.
667. Schmidt WE, Creutzfeldt W, Hocker M, Nustede R, Choudhury AR, Schleser A, 685. Sheikh SP, Holst JJ, Orskov C, Ekman R, Schwartz TW. Release of PYY from pig
Rovati LC, Folsch UR. Cholecystokinin receptor antagonist loxiglumide modulates intestinal mucosa; luminal and neural regulation. Regul Pept 26: 253–266, 1989.
plasma levels of gastro-entero-pancreatic hormones in man. Feedback control of
686. Shi YC, Hammerle CM, Lee IC, Turner N, Nguyen AD, Riepler SJ, Lin S, Sainsbury A,
cholecystokinin and gastrin secretion. Eur J Clin Invest 21: 501–511, 1991.
Herzog H, Zhang L. Adult-onset PYY overexpression in mice reduces food intake and
668. Scholtz S, Miras AD, Chhina N, Prechtl CG, Sleeth ML, Daud NM, Ismail NA, Durighel increases lipogenic capacity. Neuropeptides 46: 173–182, 2012.
G, Ahmed AR, Olbers T, Vincent RP, Alaghband-Zadeh J, Ghatei MA, Waldman AD,
687. Shi YC, Lin S, Castillo L, Aljanova A, Enriquez RF, Nguyen AD, Baldock PA, Zhang L,
Frost GS, Bell JD, le Roux CW, Goldstone AP. Obese patients after gastric bypass
Bijker MS, Macia L, Yulyaningsih E, Zhang H, Lau J, Sainsbury A, Herzog H. Peripheral-
surgery have lower brain-hedonic responses to food than after gastric banding. Gut
specific y2 receptor knockdown protects mice from high-fat diet-induced obesity.
63: 891–902, 2014.
Obesity 19: 2137–2148, 2011.
669. Schonhoff S, Baggio L, Ratineau C, Ray SK, Lindner J, Magnuson MA, Drucker DJ, 688. Shiiya T, Nakazato M, Mizuta M, Date Y, Mondal MS, Tanaka M, Nozoe S, Hosoda H,
Leiter AB. Energy homeostasis and gastrointestinal endocrine differentiation do not Kangawa K, Matsukura S. Plasma ghrelin levels in lean and obese humans and the
require the anorectic hormone peptide YY. Mol Cell Biol 25: 4189 – 4199, 2005. effect of glucose on ghrelin secretion. J Clin Endocrinol Metab 87: 240 –244, 2002.
670. Schvarcz E, Palmer M, Aman J, Horowitz M, Stridsberg M, Berne C. Physiological 689. Shikora S, Toouli J, Herrera MF, Kulseng B, Zulewski H, Brancatisano R, Kow L,
hyperglycemia slows gastric emptying in normal subjects and patients with insulin- Pantoja JP, Johnsen G, Brancatisano A, Tweden KS, Knudson MB, Billington CJ. Vagal
dependent diabetes mellitus. Gastroenterology 113: 60 – 66, 1997. blocking improves glycemic control and elevated blood pressure in obese subjects
with type 2 diabetes mellitus. J Obes 2013: 245683, 2013.
671. Schwartz GJ, McHugh PR, Moran TH. Gastric loads and cholecystokinin synergistically
stimulate rat gastric vagal afferents. Am J Physiol Regul Integr Comp Physiol 265: R872– 690. Shin AC, Berthoud HR. Food reward functions as affected by obesity and bariatric
R876, 1993. surgery. Int J Obes 35 Suppl 3: S40 – 44, 2011.
672. Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central nervous system 691. Shin AC, Zheng H, Townsend RL, Sigalet DL, Berthoud HR. Meal-induced hormone
control of food intake. Nature 404: 661– 671, 2000. responses in a rat model of Roux-en-Y gastric bypass surgery. Endocrinology 151:
1588 –1597, 2010.
673. Schwarzendrube J, Niederau M, Luthen R, Niederau C. Effects of cholecystokinin-
receptor blockade on pancreatic and biliary function in healthy volunteers. Gastroen- 692. Shrestha YB, Wickwire K, Giraudo SQ. Direct effects of nutrients, acetylcholine,
terology 100: 1683–1690, 1991. CCK, and insulin on ghrelin release from the isolated stomachs of rats. Peptides 30:
1187–1191, 2009.
674. Schwizer W, Borovicka J, Kunz P, Fraser R, Kreiss C, D’Amato M, Crelier G, Boesiger
P, Fried M. Role of cholecystokinin in the regulation of liquid gastric emptying and 693. Siegel JA, Urbain JL, Adler LP, Charkes ND, Maurer AH, Krevsky B, Knight LC, Fisher
gastric motility in humans: studies with the CCK antagonist loxiglumide. Gut 41: RS, Malmud LS. Biphasic nature of gastric emptying. Gut 29: 85– 89, 1988.
500 –504, 1997.
694. Silberbauer C, Frey-Rindova P, Langhans W. Breakfasts with different fiber and ma-
675. Sclafani A. Gut-brain nutrient signaling. Appetition vs satiation. Appetite 71: 454 – 458, cronutrient contents do not differentially affect timing, size or microstructure of the
2013. subsequent lunch. Z Ernahrungswiss 35: 356 –368, 1996.
676. Scott V, Kimura N, Stark JA, Luckman SM. Intravenous peptide YY3-36 and Y2 re- 695. Sima E, Hedberg J, Sundbom M. Gastrointestinal symptoms, weight loss and patient
ceptor antagonism in the rat: effects on feeding behaviour. J Neuroendocrinol 17: satisfaction 5 years after gastric bypass: a study of three techniques for the gastroje-
452– 457, 2005. junal anastomosis. Surg Endosc 30: 1553–1558, 2016.
696. Simonian HP, Kresge KM, Boden GH, Parkman HP. Differential effects of sham 717. Steinert RE, Gerspach AC, Gutmann H, Asarian L, Drewe J, Beglinger C. The func-
feeding and meal ingestion on ghrelin and pancreatic polypeptide levels: evidence for tional involvement of gut-expressed sweet taste receptors in glucose-stimulated se-
vagal efferent stimulation mediating ghrelin release. Neurogastroenterol Motil 17: 348 – cretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). Clin Nutr 30: 524 –
354, 2005. 532, 2011.
697. Simpson K, Parker J, Plumer J, Bloom S. CCK, PYY and PP: the control of energy 718. Steinert RE, Luscombe-Marsh ND, Little TJ, Standfield S, Otto B, Horowitz M, Feinle-
balance. Handb Exp Pharmacol 209 –230, 2012. Bisset C. Effects of intraduodenal infusion of L-tryptophan on ad libitum eating, antro-
pyloroduodenal motility, glycemia, insulinemia and gut peptide secretion in healthy
698. Sisley S, Gutierrez-Aguilar R, Scott M, D’Alessio DA, Sandoval DA, Seeley RJ. Neuro- men. J Clin Endocrinol Metab 99: 3275–32, 2014.
nal GLP1R mediates liraglutide’s anorectic but not glucose-lowering effect. J Clin Invest
124: 2456 –2463, 2014. 719. Steinert RE, Meyer-Gerspach AC, Beglinger C. The role of the stomach in the control
of appetite and the secretion of satiation peptides. Am J Physiol Endocrinol Metab 302:
699. Skibicka KP. The central GLP-1: implications for food and drug reward. Front Neurosci E666 –E673, 2012.
7: 181, 2013.
720. Steinert RE, Peterli R, Keller S, Meyer-Gerspach AC, Drewe J, Peters T, Beglinger C.
700. Skibicka KP, Shirazi RH, Hansson C, Dickson SL. Ghrelin interacts with neuropeptide Bile acids and gut peptide secretion after bariatric surgery: a 1-year prospective
Y Y1 and opioid receptors to increase food reward. Endocrinology 153: 1194 –1205, randomized pilot trial. Obesity 21: E660 –E668, 2013.
2012.
721. Steinert RE, Schirra J, Meyer-Gerspach AC, Kienle P, Fischer H, Schulte F, Goeke B,
701. Sloth B, Holst JJ, Flint A, Gregersen NT, Astrup A. Effects of PYY1-36 and PYY3-36 on Beglinger C. Effect of glucagon-like peptide-1 receptor antagonism on appetite and
appetite, energy intake, energy expenditure, glucose and fat metabolism in obese and food intake in healthy men. Am J Clin Nutr 100: 514 –523, 2014.
lean subjects. Am J Physiol Endocrinol Metab 292: E1062–E1068, 2007. 722. Stengel A, Keire D, Goebel M, Evilevitch L, Wiggins B, Tache Y, Reeve JR Jr. The
RAPID method for blood processing yields new insight in plasma concentrations and
702. Smith GP. The controls of eating: a shift from nutritional homeostasis to behavioral
molecular forms of circulating gut peptides. Endocrinology 150: 5113–5118, 2009.
neuroscience. Nutrition 16: 814 – 820, 2000.
723. Sterling P. Allostasis: a model of predictive regulation. Physiol Behav 106: 5–15, 2012.
703. Smith GP. Gut hormone hypothesis of postprandial satiety. In: Eating and Its Disorders,
edited by Stunkard AJ, Stella E. New York: Raven, 1984, p. 67–75. 724. Stevens JE, Jones KL, Rayner CK, Horowitz M. Pathophysiology and pharmacotherapy
of gastroparesis: current and future perspectives. Expert Opin Pharmacother 14: 1171–
704. Smith GP, Jerome C, Cushin BJ, Eterno R, Simansky KJ. Abdominal vagotomy blocks
1186, 2013.
the satiety effect of cholecystokinin in the rat. Science 213: 1036 –1037, 1981.
725. Stewart JE, Seimon RV, Otto B, Keast RS, Clifton PM, Feinle-Bisset C. Marked differ-
705. Smith GP, Jerome C, Norgren R. Afferent axons in abdominal vagus mediate satiety ences in gustatory and gastrointestinal sensitivity to oleic acid between lean and obese
effect of cholecystokinin in rats. Am J Physiol Regul Integr Comp Physiol 249: R638 – men. Am J Clin Nutr 93: 703–711, 2011.
R641, 1985.
726. Stice E, Yokum S, Burger KS, Epstein LH, Small DM. Youth at risk for obesity show
706. Snyder DJ, Bartoshuk LM. Epidemiological studies of taste function: discussion and greater activation of striatal and somatosensory regions to food. J Neurosci 31: 4360 –
perspectives. Ann NY Acad Sci 1170: 574 –580, 2009. 4366, 2011.
707. Spiegel K, Tasali E, Leproult R, Scherberg N, Van Cauter E. Twenty-four-hour profiles 727. Stock S, Leichner P, Wong AC, Ghatei MA, Kieffer TJ, Bloom SR, Chanoine JP.
of acylated and total ghrelin: relationship with glucose levels and impact of time of day Ghrelin, peptide YY, glucose-dependent insulinotropic polypeptide, and hunger re-
and sleep. J Clin Endocrinol Metab 96: 486 – 493, 2011. sponses to a mixed meal in anorexic, obese, and control female adolescents. J Clin
Endocrinol Metab 90: 2161–2168, 2005.
708. Spiller RC, Trotman IF, Adrian TE, Bloom SR, Misiewicz JJ, Silk DB. Further charac-
terisation of the “ileal brake” reflex in man– effect of ileal infusion of partial digests of 728. Stunkard AJ, Fox S. The relationship of gastric motility and hunger. A summary of the
fat, protein, and starch on jejunal motility and release of neurotensin, enteroglucagon, evidence. Psychosom Med 33: 123–134, 1971.
and peptide YY. Gut 29: 1042–1051, 1988.
729. Sturm K, MacIntosh CG, Parker BA, Wishart J, Horowitz M, Chapman IM. Appetite,
709. Spiller RC, Trotman IF, Higgins BE, Ghatei MA, Grimble GK, Lee YC, Bloom SR, food intake, and plasma concentrations of cholecystokinin, ghrelin, and other gastro-
Misiewicz JJ, Silk DB. The ileal brake–inhibition of jejunal motility after ileal fat perfu- intestinal hormones in undernourished older women and well-nourished young and
sion in man. Gut 25: 365–374, 1984. older women. J Clin Endocrinol Metab 88: 3747–3755, 2003.
710. Stadlbauer U, Arnold M, Weber E, Langhans W. Possible mechanisms of circulating 730. Sturm K, Parker B, Wishart J, Feinle-Bisset C, Jones KL, Chapman I, Horowitz M.
PYY-induced satiation in male rats. Endocrinology 154: 193–204, 2013. Energy intake and appetite are related to antral area in healthy young and older
subjects. Am J Clin Nutr 80: 656 – 667, 2004.
711. Starling EH. The Croonian lectures on the chemical correlation of the functions of the
body. Lancet 339 –341, 1905. 731. Stylopoulos N, Davis P, Pettit JD, Rattner DW, Kaplan LM. Changes in serum ghrelin
predict weight loss after Roux-en-Y gastric bypass in rats. Surg Endosc 19: 942–946,
712. Stefater MA, Wilson-Perez HE, Chambers AP, Sandoval DA, Seeley RJ. All bariatric 2005.
surgeries are not created equal: insights from mechanistic comparisons. Endocr Rev
732. Sugiyama K, Manaka H, Kato T, Yamatani K, Tominaga M, Sasaki H. Stimulation of
33: 595– 622, 2012.
truncated glucagon-like peptide-1 release from the isolated perfused canine ileum by
713. Steinert RE, Poller B, Castelli M, Drewe J, Beglinger C. Oral administration of gluca- glucose absorption. Digestion 55: 24 –28, 1994.
gon-like peptide 1 or peptide YY 3–36 affects food intake in healthy male subjects. Am
733. Sundaresan S, Shahid R, Riehl TE, Chandra R, Nassir F, Stenson WF, Liddle RA,
J Clin Nutr 92: 810 – 817, 2010. Abumrad NA. CD36-dependent signaling mediates fatty acid-induced gut release of
secretin and cholecystokinin. FASEB J 27: 1191–1202, 2012.
714. Steinert RE, Poller B, Castelli M, Friedman K, Huber A, Drewe J, Beglinger C. Orally
administered glucagon-like peptide-1 affects glucose homeostasis following an oral 734. Surina DM, Langhans W, Pauli R, Wenk C. Meal composition affects postprandial fatty
glucose tolerance test in healthy male subjects. Clin Pharmacol Ther 86: 644 – 650, acid oxidation. Am J Physiol Regul Integr Comp Physiol 264: R1065–R1070, 1993.
2009.
735. Suzuki S, Ramos EJ, Goncalves CG, Chen C, Meguid MM. Changes in GI hormones
715. Steinert RE, Beglinger C. Nutrient sensing in the gut: interactions between chemo- and their effect on gastric emptying and transit times after Roux-en-Y gastric bypass in
sensory cells, visceral afferents and the secretion of satiation peptides. Physiol Behav rat model. Surgery 138: 283–290, 2005.
105: 62–70, 2011.
736. Svane MS, Bojsen-Moller KN, Nielsen S, Jorgensen NB, Dirksen C, Bendtsen F,
716. Steinert RE, Frey F, Toepfer A, Drewe J, Beglinger C. Effects of carbohydrate sugars Kristiansen VB, Hartmann B, Holst JJ, Madsbad S. Effects of endogenous GLP-1 and
and artificial sweeteners on appetite and the secretion of gastrointestinal satiety GIP on glucose tolerance after Roux-en-Y gastric bypass surgery. Am J Physiol Endo-
peptides. Br J Nutr 105: 1320 –1328, 2011. crinol Metab 310: E505–E514, 2016.
737. Svane MS, Jorgensen NB, Bojsen-Moller KN, Dirksen C, Nielsen S, Kristiansen VB, 757. Theander-Carrillo C, Wiedmer P, Cettour-Rose P, Nogueiras R, Perez-Tilve D,
Torang S, Wewer Albrechtsen NJ, Rehfeld JF, Hartmann B, Madsbad S, Holst JJ. Pfluger P, Castaneda TR, Muzzin P, Schurmann A, Szanto I, Tschop MH, Rohner-
Peptide YY and glucagon-like peptide-1 contribute to decreased food intake after Jeanrenaud F. Ghrelin action in the brain controls adipocyte metabolism. J Clin Invest
Roux-en-Y gastric bypass surgery. Int J Obes (Lond). In press. 116: 1983–1993, 2006.
738. Svendsen B, Pedersen J, Albrechtsen NJ, Hartmann B, Torang S, Rehfeld JF, Poulsen 758. Theil PK, Jorgensen H, Serena A, Hendrickson J, Bach Knudsen KE. Products deriving
SS, Holst JJ. An analysis of cosecretion and coexpression of gut hormones from male from microbial fermentation are linked to insulinaemic response in pigs fed breads
rat proximal and distal small intestine. Endocrinology 156: 847– 857, 2015. prepared from whole-wheat grain and wheat and rye ingredients. Br J Nutr 105:
373–383, 2011.
739. Swartz TD, Savastano DM, Covasa M. Reduced sensitivity to cholecystokinin in male
rats fed a high-fat diet is reversible. J Nutr 140: 1698 –1703, 2010. 759. Theodorakis MJ, Carlson O, Michopoulos S, Doyle ME, Juhaszova M, Petraki K, Egan
JM. Human duodenal enteroendocrine cells: source of both incretin peptides, GLP-1
740. Sweeney TE, Morton JM. Metabolic surgery: action via hormonal milieu changes, and GIP. Am J Physiol Endocrinol Metab 290: E550 –E559, 2006.
changes in bile acids or gut microbiota? A summary of the literature. Best Pract Res Clin
Gastroenterol 28: 727–740, 2014. 760. Thimister PW, Hopman WP, Sloots CE, Rosenbusch G, Willems HL, Trijbels FJ,
Jansen JB. Role of intraduodenal proteases in plasma cholecystokinin and pancreati-
741. Swinburn B, Sacks G, Ravussin E. Increased food energy supply is more than sufficient cobiliary responses to protein and amino acids. Gastroenterology 110: 567–575, 1996.
to explain the US epidemic of obesity. Am J Clin Nutr 90: 1453–1456, 2009.
761. Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, Macchiarulo A, Yamamoto H,
742. Sykaras AG, Demenis C, Cheng L, Pisitkun T, McLaughlin JT, Fenton RA, Smith CP. Mataki C, Pruzanski M, Pellicciari R, Auwerx J, Schoonjans K. TGR5-mediated bile acid sensing
Duodenal CCK cells from male mice express multiple hormones including ghrelin. controls glucose homeostasis. Cell Metab 10: 167–177, 2009.
Endocrinology 155: 3339 –3351, 2014.
762. Thomsen C, Rasmussen O, Lousen T, Holst JJ, Fenselau S, Schrezenmeir J, Herman-
743. Sysko R, Devlin MJ, Walsh BT, Zimmerli E, Kissileff HR. Satiety and test meal intake sen K. Differential effects of saturated and monounsaturated fatty acids on postpran-
among women with binge eating disorder. Int J Eat Disord 40: 554 –561, 2007. dial lipemia and incretin responses in healthy subjects. Am J Clin Nutr 69: 1135–1143,
1999.
744. Szarka LA, Camilleri M. Methods for measurement of gastric motility. Am J Physiol
Gastrointest Liver Physiol 296: G461–G475, 2009. 763. Toft-Nielsen MB, Damholt MB, Madsbad S, Hilsted LM, Hughes TE, Michelsen BK,
Holst JJ. Determinants of the impaired secretion of glucagon-like peptide-1 in type 2
745. Tack J, Deloose E. Complications of bariatric surgery: dumping syndrome, reflux and diabetic patients. J Clin Endocrinol Metab 86: 3717–3723, 2001.
vitamin deficiencies. Best Pract Res Clin Gastroenterol 28: 741–749, 2014.
764. Toft-Nielsen MB, Madsbad S, Holst JJ. Determinants of the effectiveness of glucagon-
746. Tack J, Deloose E, Ang D, Scarpellini E, Vanuytsel T, Van Oudenhove L, Depoortere like peptide-1 in type 2 diabetes. J Clin Endocrinol Metab 86: 3853–3860, 2001.
I. Motilin-induced gastric contractions signal hunger in man. Gut 65: 214 –224, 2016.
765. Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, Cameron J,
747. Tack J, Demedts I, Meulemans A, Schuurkes J, Janssens J. Role of nitric oxide in the Grosse J, Reimann F, Gribble FM. Short-chain fatty acids stimulate glucagon-like
gastric accommodation reflex and in meal induced satiety in humans. Gut 51: 219 – peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 61: 364 –
224, 2002. 371, 2012.
748. Tack J, Depoortere I, Bisschops R, Verbeke K, Janssens J, Peeters T. Influence of 766. Tomas E, Habener JF. Insulin-like actions of glucagon-like peptide-1: a dual receptor
ghrelin on gastric emptying and meal-related symptoms in idiopathic gastroparesis. hypothesis. Trends Endocrinol Metab 21: 59 – 67, 2010.
Aliment Pharmacol Ther 22: 847– 853, 2005.
767. Tong J, Prigeon RL, Davis HW, Bidlingmaier M, Tschop MH, D’Alessio D. Physiologic
749. Tai K, Hammond AJ, Wishart JM, Horowitz M, Chapman IM. Carbohydrate and fat concentrations of exogenously infused ghrelin reduces insulin secretion without af-
digestion is necessary for maximal suppression of total plasma ghrelin in healthy adults. fecting insulin sensitivity in healthy humans. J Clin Endocrinol Metab 98: 2536 –2543,
Appetite 55: 407– 412, 2010. 2013.
768. Torang S, Bojsen-Moller KN, Svane MS, Hartmann B, Rosenkilde MM, Madsbad S,
750. Takaya K, Ariyasu H, Kanamoto N, Iwakura H, Yoshimoto A, Harada M, Mori K,
Holst JJ. In vivo and in vitro degradation of peptide YY3-36 to inactive peptide YY3-34
Komatsu Y, Usui T, Shimatsu A, Ogawa Y, Hosoda K, Akamizu T, Kojima M, Kangawa
in humans. Am J Physiol Regul Integr Comp Physiol 310: R866 –R874, 2016.
K, Nakao K. Ghrelin strongly stimulates growth hormone release in humans. J Clin
Endocrinol Metab 85: 4908 – 4911, 2000. 769. Torang S, Veedfald S, Rosenkilde MM, Hartmann B, Holst JJ. The anorexic hormone
Peptide YY3-36 is rapidly metabolized to inactive Peptide YY3-34 in vivo. Physiol Rep
751. Takei I, Miyamoto K, Funae O, Ohashi N, Meguro S, Tokui M, Saruta T. Secretion of
3: 2015.
GIP in responders to acarbose in obese Type 2(NIDDM) patients. J Diabetes Compli-
cations 15: 245–249, 2001. 770. Torra S, Ilzarbe L, Malagelada JR, Negre M, Mestre-Fusco A, Aguade-Bruix S, Florensa
E, Sune P, Gras B, Hernandez JJ, Casamitjana R, Garcia MA, Ros FB, Delgado-Aros S.
752. Tanaka T, Katsuma S, Adachi T, Koshimizu Ta Hirasawa A, Tsujimoto G. Free fatty
Meal size can be decreased in obese subjects through pharmacological acceleration of
acids induce cholecystokinin secretion through GPR120. Naunyn-Schmiedebergs Arch
gastric emptying (The OBERYTH trial). Int J Obes (Lond) 35: 829 – 837, 2011.
Pharmacol 377: 523–527, 2008.
771. Tosh SM. Review of human studies investigating the post-prandial blood-glucose
753. Tarini J, Wolever TM. The fermentable fibre inulin increases postprandial serum lowering ability of oat and barley food products. Eur J Clin Nutr 67: 310 –317, 2013.
short-chain fatty acids and reduces free-fatty acids and ghrelin in healthy subjects. Appl
Physiol Nutr Metab 35: 9 –16, 2010. 772. Tougas G, Eaker EY, Abell TL, Abrahamsson H, Boivin M, Chen J, Hocking MP, Quigley EM,
Koch KL, Tokayer AZ, Stanghellini V, Chen Y, Huizinga JD, Ryden J, Bourgeois I, McCallum
754. Tazoe H, Otomo Y, Kaji I, Tanaka R, Karaki SI, Kuwahara A. Roles of short-chain fatty RW. Assessment of gastric emptying using a low fat meal: establishment of international control
acids receptors, GPR41 and GPR43 on colonic functions. J Physiol Pharmacol 59 Suppl values. Am J Gastroenterol 95: 1456–1462, 2000.
2: 251–262, 2008.
773. Trabelsi MS, Daoudi M, Prawitt J, Ducastel S, Touche V, Sayin SI, Perino A, Brighton
755. Teff KL, Elliott SS, Tschop M, Kieffer TJ, Rader D, Heiman M, Townsend RR, Keim CA, Sebti Y, Kluza J, Briand O, Dehondt H, Vallez E, Dorchies E, Baud G, Spinelli V,
NL, D’Alessio D, Havel PJ. Dietary fructose reduces circulating insulin and leptin, Hennuyer N, Caron S, Bantubungi K, Caiazzo R, Reimann F, Marchetti P, Lefebvre P,
attenuates postprandial suppression of ghrelin, and increases triglycerides in women. Backhed F, Gribble FM, Schoonjans K, Pattou F, Tailleux A, Staels B, Lestavel S.
J Clin Endocrinol Metab 89: 2963–2972, 2004. Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine
L cells. Nat Commun 6: 7629, 2015.
756. Thazhath SS, Marathe CS, Wu T, Chang J, Khoo J, Kuo P, Checklin HL, Bound MJ,
Rigda RS, Crouch B, Jones KL, Horowitz M, Rayner CK. The glucagon-like peptide 1 774. Trahair LG, Horowitz M, Rayner CK, Gentilcore D, Lange K, Wishart JM, Jones KL.
receptor agonist exenatide inhibits small intestinal motility, flow, transit, and absorp- Comparative effects of variations in duodenal glucose load on glycemic, insulinemic,
tion of glucose in healthy subjects and patients with type 2 diabetes: a randomized and incretin responses in healthy young and older subjects. J Clin Endocrinol Metab 97:
controlled trial. Diabetes 65: 269 –275, 2016. 844 – 851, 2012.
775. Trevaskis JL, Parkes DG, Roth JD. Insights into amylin-leptin synergy. Trends Endocri- 796. Verdich C, Flint A, Gutzwiller JP, Naslund E, Beglinger C, Hellstrom PM, Long SJ,
nol Metab 21: 473– 479, 2010. Morgan LM, Holst JJ, Astrup A. 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:
776. Trevaskis JL, Sun C, Athanacio J, D’Souza L, Samant M, Tatarkiewicz K, Griffin PS, 4382– 4389, 2001.
Wittmer C, Wang Y, Teng CH, Forood B, Parkes DG, Roth JD. Synergistic metabolic
benefits of an exenatide analogue and cholecystokinin in diet-induced obese and 797. Verdich C, Madsen JL, Toubro S, Buemann B, Holst JJ, Astrup A. Effect of obesity and
leptin-deficient rodents. Diabetes Obes Metab 17: 61–73, 2015. major weight reduction on gastric emptying. Int J Obes Relat Metab Disord 24: 899 –
905, 2000.
777. Trevaskis JL, Turek VF, Griffin PS, Wittmer C, Parkes DG, Roth JD. Multi-hormonal
weight loss combinations in diet-induced obese rats: therapeutic potential of chole- 798. Verdich C, Toubro S, Buemann B, Lysgard Madsen J, Juul Holst J, Astrup A. The role
cystokinin? Physiol Behav 100: 187–195, 2010. of postprandial releases of insulin and incretin hormones in meal-induced satiety–
effect of obesity and weight reduction. Int J Obes Relat Metab Disord 25: 1206 –1214,
778. Tschop M, Wawarta R, Riepl RL, Friedrich S, Bidlingmaier M, Landgraf R, Folwaczny 2001.
C. Post-prandial decrease of circulating human ghrelin levels. J Endocrinol Invest 24:
RC19 –21, 2001. 799. Verhulst PJ, Depoortere I. Ghrelin’s second life: from appetite stimulator to glucose
regulator. World J Gastroenterol 18: 3183–3195, 2012.
779. Tschop M, Weyer C, Tataranni PA, Devanarayan V, Ravussin E, Heiman ML. Circu-
lating ghrelin levels are decreased in human obesity. Diabetes 50: 707–709, 2001. 800. Vestergaard ET, Hansen TK, Gormsen LC, Jakobsen P, Moller N, Christiansen JS,
Jorgensen JO. Constant intravenous ghrelin infusion in healthy young men: clinical
780. Uchida A, Zechner JF, Mani BK, Park WM, Aguirre V, Zigman JM. Altered ghrelin
pharmacokinetics and metabolic effects. Am J Physiol Endocrinol Metab 292: E1829 –
secretion in mice in response to diet-induced obesity and Roux-en-Y gastric bypass.
E1836, 2007.
Mol Metab 3: 717–730, 2014.
801. Viardot A, Heilbronn LK, Herzog H, Gregersen S, Campbell LV. Abnormal postpran-
781. Umapathysivam MM, Lee MY, Jones KL, Annink CE, Cousins CE, Trahair LG, Rayner
dial PYY response in insulin sensitive nondiabetic subjects with a strong family history
CK, Chapman MJ, Nauck MA, Horowitz M, Deane AM. Comparative effects of pro-
of type 2 diabetes. Int J Obes (Lond) 32: 943–948, 2008.
longed and intermittent stimulation of the glucagon-like peptide-1 receptor on gastric
emptying and glycaemia. Diabetes 63: 785–790, 2014. 802. Villar HV, Wangenteen SL, Burks TF, Patton DD. Mechanisms of satiety and gastric
emptying after gastric partitioning and bypass. Surgery 90: 229 –236, 1981.
782. Unger RH, Eisentraut AM. Entero-insular axis. Arch Intern Med 123: 261–266, 1969.
803. Vilsboll T, Krarup T, Madsbad S, Holst JJ. Both GLP-1 and GIP are insulinotropic at
783. Ussher JR, Drucker DJ. Cardiovascular biology of the incretin system. Endocr Rev 33:
basal and postprandial glucose levels and contribute nearly equally to the incretin
187–215, 2012.
effect of a meal in healthy subjects. Regul Pept 114: 115–121, 2003.
784. Uttenthal LO, Ukponmwan OO, Ghiglione M, Bloom SR. Acute and short term
804. Vilsboll T, Krarup T, Sonne J, Madsbad S, Volund A, Juul AG, Holst JJ. Incretin secretion
effects of intestinal alpha-glucosidase inhibition on gut hormone responses in man. Dig
in relation to meal size and body weight in healthy subjects and people with type 1 and
Dis Sci 32: 139 –144, 1987.
type 2 diabetes mellitus. J Clin Endocrinol Metab 88: 2706 –2713, 2003.
785. Uttenthal LO, Ukponmwan OO, Wood SM, Ghiglione M, Ghatei MA, Trayner IM,
805. Vollmer K, Holst JJ, Baller B, Ellrichmann M, Nauck MA, Schmidt WE, Meier JJ.
Bloom SR. Long-term effects of intestinal alpha-glucosidase inhibition on postprandial
glucose, pancreatic and gut hormone responses and fasting serum lipids in diabetics on Predictors of incretin concentrations in subjects with normal, impaired, and diabetic
sulphonylureas. Diabet Med 3: 155–160, 1986. glucose tolerance. Diabetes 57: 678 – 687, 2008.
786. Vahl TP, Drazen DL, Seeley RJ, D’Alessio DA, Woods SC. Meal-anticipatory glucagon- 806. Vrang N, Phifer CB, Corkern MM, Berthoud HR. Gastric distension induces c-Fos in
like peptide-1 secretion in rats. Endocrinology 151: 569 –575, 2010. medullary GLP-1/2-containing neurons. Am J Physiol Regul Integr Comp Physiol 285:
R470 –R478, 2003.
787. Van Avesaat M, Troost FJ, Ripken D, Hendriks HF, Masclee AA. Ileal brake activation:
macronutrient-specific effects on eating behavior? Int J Obes (Lond) 39: 235–243, 807. Walther C, Morl K, Beck-Sickinger AG. Neuropeptide Y receptors: ligand binding and
2015. trafficking suggest novel approaches in drug development. J Pept Sci 17: 233–246,
2011.
788. Van Bloemendaal L, IJzerman RG, Ten Kulve JS, Barkhof F, Konrad RJ, Drent ML,
Veltman DJ, Diamant M. GLP-1 receptor activation modulates appetite- and reward- 808. Wang G, Agenor K, Pizot J, Kotler DP, Harel Y, Van Der Schueren BJ, Quercia I,
related brain areas in humans. Diabetes 63: 4186 – 4196, 2014. McGinty J, Laferrere B. Accelerated gastric emptying but no carbohydrate malabsorp-
tion 1 year after gastric bypass surgery (GBP). Obes Surg 22: 1263–1267, 2012.
789. Van Can J, Sloth B, Jensen CB, Flint A, Blaak EE, Saris WH. Effects of the once-daily
GLP-1 analog liraglutide on gastric emptying, glycemic parameters, appetite and en- 809. Wang GJ, Tomasi D, Backus W, Wang R, Telang F, Geliebter A, Korner J, Bauman A,
ergy metabolism in obese, non-diabetic adults. Int J Obes (Lond) 38: 784 –793, 2014. Fowler JS, Thanos PK, Volkow ND. Gastric distention activates satiety circuitry in the
human brain. Neuroimage 39: 1824 –1831, 2008.
790. Van den Hoek AM, Heijboer AC, Corssmit EP, Voshol PJ, Romijn JA, Havekes LM, Pijl
H. PYY3-36 reinforces insulin action on glucose disposal in mice fed a high-fat diet. 810. Wang Q, Liu C, Uchida A, Chuang JC, Walker A, Liu T, Osborne-Lawrence S, Mason
Diabetes 53: 1949 –1952, 2004. BL, Mosher C, Berglund ED, Elmquist JK, Zigman JM. Arcuate AgRP neurons mediate
orexigenic and glucoregulatory actions of ghrelin. Mol Metab 3: 64 –72, 2014.
791. Vandenbergh J, Dupont P, Fischler B, Bormans G, Persoons P, Janssens J, Tack J.
Regional brain activation during proximal stomach distention in humans: a positron 811. Wang Y, Chandra R, Samsa LA, Gooch B, Fee BE, Cook JM, Vigna SR, Grant AO,
emission tomography study. Gastroenterology 128: 564 –573, 2005. Liddle RA. Amino acids stimulate cholecystokinin release through the Ca2⫹-sensing
receptor. Am J Physiol Gastrointest Liver Physiol 300: G528 –G537, 2011.
792. Vandevijvere S, Chow CC, Hall KD, Umali E, Swinburn BA. Increased food energy
supply as a major driver of the obesity epidemic: a global analysis. Bull World Health 812. Wang Y, Prpic V, Green GM, Reeve JR Jr, Liddle RA. Luminal CCK-releasing factor
Organ 93: 446 – 456, 2015. stimulates CCK release from human intestinal endocrine and STC-1 cells. Am J Physiol
Gastrointest Liver Physiol 282: G16 –G22, 2002.
793. Vanis L, Hausken T, Gentilcore D, Rigda RS, Rayner CK, Feinle-Bisset C, Horowitz M,
Jones KL. Comparative effects of glucose and xylose on blood pressure, gastric emp- 813. Wansink B. From mindless eating to mindlessly eating better. Physiol Behav 100:
tying and incretin hormones in healthy older subjects. Br J Nutr 105: 1644 –1651, 454 – 463, 2010.
2011.
814. Washington MC, Williams K, Sayegh AI. The feeding responses evoked by endoge-
794. Vazquez Roque MI, Camilleri M, Stephens DA, Jensen MD, Burton DD, Baxter KL, nous cholecystokinin are regulated by different gastrointestinal sites. Horm Behav 78:
Zinsmeister AR. Gastric sensorimotor functions and hormone profile in normal 79 – 85, 2015.
weight, overweight, and obese people. Gastroenterology 131: 1717–1724, 2006.
815. Wasse LK, Sunderland C, King JA, Batterham RL, Stensel DJ. Influence of rest and exercise
795. Velchik MG, Reynolds JC, Alavi A. The effect of meal energy content on gastric at a simulated altitude of 4,000 m on appetite, energy intake, and plasma concentrations of
emptying. J Nucl Med 30: 1106 –1110, 1989. acylated ghrelin and peptide YY. J Appl Physiol (1985) 112: 552–559, 2012.
816. Waters MJ. Endocrinology: the next 60 years–the helix and the chip. J Endocrinol 190: 838. Wortley KE, Garcia K, Okamoto H, Thabet K, Anderson KD, Shen V, Herman JP,
11–12, 2006. Valenzuela D, Yancopoulos GD, Tschop MH, Murphy A, Sleeman MW. Peptide YY
regulates bone turnover in rodents. Gastroenterology 133: 1534 –1543, 2007.
817. Wehrwein EA, Carter JR. The mind matters: psychology as an overlooked variable
within physiology studies. Physiology 31: 74 –75, 2016. 839. Wren AM, Seal LJ, Cohen MA, Brynes AE, Frost GS, Murphy KG, Dhillo WS, Ghatei
MA, Bloom SR. Ghrelin enhances appetite and increases food intake in humans. J Clin
818. Weise CM, Thiyyagura P, Reiman EM, Chen K, Krakoff J. Postprandial plasma PYY Endocrinol Metab 86: 5992, 2001.
concentrations are associated with increased regional gray matter volume and rCBF
declines in caudate nuclei–a combined MRI and H2(15)O PET study. Neuroimage 60: 840. Wu T, Bound MJ, Standfield SD, Gedulin B, Jones KL, Horowitz M, Rayner CK. Effects
592– 600, 2012. of rectal administration of taurocholic acid on glucagon-like peptide-1 and peptide YY
secretion in healthy humans. Diabetes Obes Metab 15: 474 – 477, 2012.
819. Wen J, Phillips SF, Sarr MG, Kost LJ, Holst JJ. PYY and GLP-1 contribute to feedback
inhibition from the canine ileum and colon. Am J Physiol Gastrointest Liver Physiol 269: 841. Wu T, Bound MJ, Standfield SD, Jones KL, Horowitz M, Rayner CK. Effects of tauro-
G945–G952, 1995. cholic acid on glycemic, glucagon-like peptide-1, and insulin responses to small intes-
tinal glucose infusion in healthy humans. J Clin Endocrinol Metab 98: E718 –722, 2013.
820. Werling M, Vincent RP, Cross GF, Marschall HU, Fandriks L, Lonroth H, Taylor DR,
Alaghband-Zadeh J, Olbers T, Le Roux CW. Enhanced fasting and post-prandial 842. Wu T, Ma J, Bound MJ, Checklin H, Deacon CF, Jones KL, Horowitz M, Rayner CK.
plasma bile acid responses after Roux-en-Y gastric bypass surgery. Scand J Gastroen- Effects of sitagliptin on glycemia, incretin hormones, and antropyloroduodenal motil-
terol 48: 1257–1264, 2013. ity in response to intraduodenal glucose infusion in healthy lean and obese humans,
and patients with type 2 diabetes treated with or without metformin. Diabetes 63:
821. West DB, Fey D, Woods SC. Cholecystokinin persistently suppresses meal size but 2776 –2787, 2014.
not food intake in free-feeding rats. Am J Physiol Regul Integr Comp Physiol 246: R776 –
R787, 1984. 843. Wu T, Rayner CK, Young RL, Horowitz M. Gut motility and enteroendocrine secre-
tion. Curr Opin Pharmacol 13: 928 –934, 2013.
822. Wettergren A, Schjoldager B, Mortensen PE, Myhre J, Christiansen J, Holst JJ. Trun-
cated GLP-1 (proglucagon 78 –107-amide) inhibits gastric and pancreatic functions in 844. Wu T, Thazhath SS, Marathe CS, Bound MJ, Jones KL, Horowitz M, Rayner CK.
man. Dig Dis Sci 38: 665– 673, 1993. Comparative effect of intraduodenal and intrajejunal glucose infusion on the gut-
incretin axis response in healthy males. Nutr Diabetes 5: e156, 2015.
823. Wickbom J, Herrington MK, Permert J, Jansson A, Arnelo U. Gastric emptying in
response to IAPP and CCK in rats with subdiaphragmatic afferent vagotomy. Regul 845. Wu T, Zhao BR, Bound MJ, Checklin HL, Bellon M, Little TJ, Young RL, Jones KL,
Pept 148: 21–25, 2008. Horowitz M, Rayner CK. Effects of different sweet preloads on incretin hormone
secretion, gastric emptying, and postprandial glycemia in healthy humans. Am J Clin
824. Widmayer P, Breer H, Hass N. Candidate chemosensory cells in the porcine stomach. Nutr 95: 78 – 83, 2012.
Histochem Cell Biol 136: 37– 45, 2011.
846. Yan W, Polidori D, Yieh L, Di J, Wu X, Moreno V, Li L, Briscoe CP, Shankley N, Dohm
825. Widmayer P, Kuper M, Kramer M, Konigsrainer A, Breer H. Altered expression of
GL, Pories WJ. Effects of meal size on the release of GLP-1 and PYY after Roux-en-Y
gustatory-signaling elements in gastric tissue of morbidly obese patients. Int J Obes
gastric bypass surgery in obese subjects with or without type 2 diabetes. Obes Surg 24:
(Lond) 36: 1353–1359, 2012.
1969 –1974, 2014.
826. Wierup N, Bjorkqvist M, Westrom B, Pierzynowski S, Sundler F, Sjolund K. Ghrelin
847. Yang J, Brown MS, Liang G, Grishin NV, Goldstein JL. Identification of the acyltrans-
and motilin are cosecreted from a prominent endocrine cell population in the small
ferase that octanoylates ghrelin, an appetite-stimulating peptide hormone. Cell 132:
intestine. J Clin Endocrinol Metab 92: 3573–3581, 2007.
387–396, 2008.
827. Williams DL. Neural integration of satiation and food reward: role of GLP-1 and
848. Ye J, Hao Z, Mumphrey MB, Townsend RL, Patterson LM, Stylopoulos N, Munzberg
orexin pathways. Physiol Behav 136: 194 –199, 2014.
H, Morrison CD, Drucker DJ, Berthoud HR. GLP-1 receptor signaling is not required
828. Williams DL, Baskin DG, Schwartz MW. Evidence that intestinal glucagon-like pep- for reduced body weight after RYGB in rodents. Am J Physiol Regul Integr Comp Physiol
tide-1 plays a physiological role in satiety. Endocrinology 150: 1680 –1687, 2009. 306: R352–R362, 2014.
829. Williams DL, Cummings DE, Grill HJ, Kaplan JM. Meal-related ghrelin suppression 849. Young RL. Sensing via intestinal sweet taste pathways. Front Neurosci 5: 23, 2011.
requires postgastric feedback. Endocrinology 144: 2765–2767, 2003.
850. Zhang J, Ritter RC. Circulating GLP-1 and CCK-8 reduce food intake by capsaicin-
830. Williams DL, Grill HJ, Cummings DE, Kaplan JM. Vagotomy dissociates short- and insensitive, nonvagal mechanisms. Am J Physiol Regul Integr Comp Physiol 302: R264 –
long-term controls of circulating ghrelin. Endocrinology 144: 5184 –5187, 2003. R273, 2012.
831. Williams DL, Hyvarinen N, Lilly N, Kay K, Dossat A, Parise E, Torregrossa AM. 851. Zhang L, Riepler SJ, Turner N, Enriquez RF, Lee IC, Baldock PA, Herzog H, Sainsbury
Maintenance on a high-fat diet impairs the anorexic response to glucagon-like-pep- A. Y2 and Y4 receptor signaling synergistically act on energy expenditure and physical
tide-1 receptor activation. Physiol Behav 103: 557–564, 2011. activity. Am J Physiol Regul Integr Comp Physiol 299: R1618 –R1628, 2010.
832. Williams KE, Washington MC, Johnson-Rouse T, Johnson RE, Freeman C, Reed C, 852. Zhao TJ, Sakata I, Li RL, Liang G, Richardson JA, Brown MS, Goldstein JL, Zigman JM.
Heath J, Sayegh AI. Exogenous glucagon-like peptide-1 acts in sites supplied by the Ghrelin secretion stimulated by {beta}1-adrenergic receptors in cultured ghrelinoma
cranial mesenteric artery to reduce meal size and prolong the intermeal interval in cells and in fasted mice. Proc Natl Acad Sci USA 107: 15868 –15873, 2010.
rats. Appetite 96: 254 –259, 2015.
853. Zheng J, Ariga H, Taniguchi H, Ludwig K, Takahashi T. Ghrelin regulates gastric phase
833. Wilson JD. The evolution of endocrinology. Clin Endocrinol 62: 389 –396, 2005. III-like contractions in freely moving conscious mice. Neurogastroenterol Motil 21:
78 – 84, 2009.
834. Witte AB, Gryback P, Holst JJ, Hilsted L, Hellstrom PM, Jacobsson H, Schmidt PT.
Differential effect of PYY1-36 and PYY3-36 on gastric emptying in man. Regul Pept 854. Zhou D, Jiang X, Ding W, Zhang D, Yang L, Zhen C, Lu L. Impact of bariatric surgery
158: 57– 62, 2009. on ghrelin and obestatin levels in obesity or type 2 diabetes mellitus rat model. J
Diabetes Res 2014: 569435, 2014.
835. Woerle HJ, Carneiro L, Derani A, Goke B, Schirra J. The role of endogenous incretin
secretion as amplifier of glucose-stimulated insulin secretion in healthy subjects and 855. Ziessman HA, Fahey FH, Atkins FB, Tall J. Standardization and quantification of ra-
patients with type 2 diabetes. Diabetes 61: 2349 –2358, 2012. dionuclide solid gastric-emptying studies. J Nucl Med 45: 760 –764, 2004.
836. Woods SC. Gastrointestinal satiety signals. I. An overview of gastrointestinal signals 856. Zwirska-Korczala K, Konturek SJ, Sodowski M, Wylezol M, Kuka D, Sowa P, Adam-
that influence food intake. Am J Physiol Gastrointest Liver Physiol 286: G7–G13, 2004. czyk-Sowa M, Kukla M, Berdowska A, Rehfeld JF, Bielanski W, Brzozowski T. Basal
and postprandial plasma levels of PYY, ghrelin, cholecystokinin, gastrin and insulin in
837. Woods SC, D’Alessio DA. Central control of body weight and appetite. J Clin Endo- women with moderate and morbid obesity and metabolic syndrome. J Physiol Phar-
crinol Metab 93: S37–50, 2008. macol 58 Suppl 1: 13–35, 2007.