You are on page 1of 5

REVIEW

CURRENT
OPINION Cholecystokinin
Graham J. Dockray

Purpose of review
Cholecystokinin (CCK) controls nutrient delivery to the small intestine by inhibiting food intake and gastric
emptying. This review deals with recent work shedding new light on how and when.
Recent findings
Intestinal I-cells release CCK in response to dietary lipid and protein through mechanisms involving the
G-protein-coupled receptors GPR40 and calcium-sensing receptor. Vagal afferent neurons are a primary
target of CCK and are now recognized as an important site of integration of peripheral signals regulating
ingestion. In addition to regulating vagal afferent nerve discharge, CCK also controls the expression of
receptors and peptide neurotransmitters by these neurons; these actions are potentiated by leptin and
inhibited by ghrelin. The responses of vagal afferent neurons to CCK are attenuated in obesity. Studies of
human central nervous system responses using functional magnetic resonance imaging indicate activation
of brainstem, hypothalamus and motor cortex by ingested fatty acid that is inhibited by a CCK-1 receptor
antagonist. CCK may also play a role in adaptive responses in pancreatic islets by maintaining b-cell mass
and acting as an incretin in certain circumstances.
Summary
CCK mediates inhibition of food intake in response to ingested lipid and protein; resistance to CCK occurs
in obesity and may contribute to altered mechanisms regulating food intake.
Keywords
I-cell, obesity, satiety, vagal afferent neurons

INTRODUCTION smooth muscle, but vagal afferent neurons are an


Ingestion of food activates a neuro-endocrine pro- important target for control of food intake and
gram directed at matching delivery of nutrients to gastric emptying. In the last year or so, progress
the small intestine with the capacity for their diges- has been made in understanding the mechanisms
tion, absorption and assimilation. The program of CCK release, its actions on vagal afferent neurons
includes sensing mechanisms for nutrients and ot- and the changes in signalling that occur in obesity.
her food components in the gut, signalling to the Several recent reviews have dealt with different
central nervous system to regulate ingestive behav- aspects of the biology of this system [1–5].
iour and autonomic efferent pathways, control of
secretion by the stomach and pancreas, gallbladder
RELEASE MECHANISMS
contraction, control of nutrient mixing and pro-
gression along the gut by changes in motility and Fatty acids and protein in the small intestine have
signalling to the endocrine pancreas to activate been recognized for over 60 years to stimulate CCK
hormonal mechanisms regulating nutrient utiliz- release, probably by acting directly on intestinal
ation. An impressive diversity of humoral mediators
has been implicated in these events and there are
Physiological Laboratory, Institute of Translational Medicine, University of
many examples of overlapping functions. In recent
Liverpool, Liverpool, UK
years, however, cholecystokinin (CCK) has emerged
Correspondence to Graham J. Dockray, Physiological Laboratory, Insti-
as an important integrator of upper small intestinal tute of Translational Medicine, University of Liverpool, Crown Street, P.O.
function that acts by inhibiting food intake and Box 147, Liverpool L69 3BX, UK. Tel: +44 151 794 5324;
gastric emptying and by stimulating pancreatic e-mail: g.j.dockray@liverpool.ac.uk
enzyme secretion and gallbladder contraction. It
is well established that there are direct effects of Curr Opin Endocrinol Diabetes Obes 2012, 19:8–12
CCK on pancreatic acinar cells and gallbladder DOI:10.1097/MED.0b013e32834eb77d

www.co-endocrinology.com Volume 19  Number 1  February 2012

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Cholecystokinin Dockray

inhibition of gastric emptying by several hexoses


KEY POINTS was reduced by a CCK-1 receptor antagonist,
 CCK is released by long-chain fatty acids and aromatic suggesting a role for CCK in carbohydrate regulation
amino acids via GPR40 and calcium-sensing receptor. of gastric motility, perhaps by providing a back-
ground tone of vagal afferent stimulation [10].
 fMRI studies in humans indicate that ingestion of lipid
activates hypothalamus and brain stem mainly by
release of CCK. NEURAL ACTIONS OF CHOLECYSTOKININ
 Vagal afferent neurons are a key target of CCK which There has been recent progress in elucidating the
stimulates expression in these neurons of cocaine and actions of CCK both on the human brain and on
amphetamine regulated transcript protein (CARTp) and vagal, brain stem and hypothalamic neurons in
Y2 receptors and inhibits expression of melanin
experimental animals.
concentrating hormone and cannabinoid receptor-1
(CB1) receptors.
 Leptin potentiates the action of CCK on vagal afferent Human central nervous system responses to
neurons; resistance to CCK and leptin in these neurons cholecystokinin
is an early feature of diet-induced obesity.
Elucidation of the mechanisms underlying central
 Studies in mice null for CCK or the CCK-1 receptor nervous system (CNS) responses to circulating CCK
reveal novel roles in adaptive mechanisms in the islets has been the subject of intense interest. Studies over
of Langerhans and in vagal afferent signalling. many years have contributed to the idea that CCK
released from the small intestine acts directly on
vagal afferent neurons that terminate in the nucleus
I-cells. The cellular mechanisms are now becoming tractus solitarius (NTS) and activate ascending path-
clearer. The availability of transgenic mice express- ways that control ingestive behaviour. The matrix of
ing green fluorescent protein (GFP) under the con- human CNS responses to endogenous CCK has now
trol of the CCK gene promoter has facilitated the been characterized by functional magnetic resonance
preparation of highly enriched populations of CCK- imaging (fMRI) following intragastric administration
expressing I-cells using fluorescence-activated cell of dodecanoate with or without the CCK-1 receptor
sorting. This approach has identified expression in &&
antagonist, dexloxiglumide [11 ]. In response to a
I-cells of GPR40 which is a G-protein-coupled recep- relatively low dose of dodecanoate, there was acti-
tor responding to long-chain fatty acids. Agonists of vation of the brainstem and hypothalamus, and also
GPR40 such as linoleic acid increased intracellular of the motor cortex; the responses were virtually
calcium and CCK release in GFP-labelled cells, abolished by dexloxiglumide, indicating the import-
whereas in cells prepared from GPR40/ mice both ance of CCK as a mediator of human CNS responses to
responses were inhibited; moreover, in vivo, the ingested fatty acids.
increase in CCK secretion in response to olive oil
was reduced by about 40% in GPR40/ mice [6 ]. A
&

similar approach has indicated a role for the extra- Vagal responses
cellular calcium-sensing receptor (CaSR) in media- The CCK-1 receptor mediates the action of CCK in
ting the effect of aromatic amino acids on CCK stimulating the discharge of vagal afferent neurons
release. Two groups have identified CaSR expression and increasing intracellular calcium concentrations.
in GFP-labelled I-cells, and have shown that aro- Vagal afferent neurons serving both the stomach
matic amino acids stimulate intracellular calcium and small intestine are activated by CCK, but their
fluxes via a CaSR-dependent mechanism [7,8]. In relative roles have been unclear. Recordings of
addition, the release of CCK in response to a pep- different populations of vagal afferent and efferent
tone meal was reduced in CaSR-null cells [7]. nerve fibres serving the proximal and distal parts of
The role of CCK in mediating the effects of the rat stomach and small intestine support the idea
ingested carbohydrate remains uncertain. The sweet that CCK acts initially on intestinal afferent fibres,
taste receptors, T1R2/T1R3, are poorly expressed by probably via a paracrine mechanism, to inhibit an
I-cells, although there is strong expression in other excitatory vagal efferent pathway to the distal
entero-endocrine cells, notably the L-cells that stomach; subsequently, there is stimulation of gas-
secrete glucagon-like peptide (GLP)-1 and peptide tric vagal afferents, corresponding to a hormonal
tyrosine tyrosine (PYY). Consistent with this, the effect, that is coupled to stimulation of an inhibitory
T1R2/T1R3 inhibitor lactisole reduced glucose- vagal efferent pathway to the proximal stomach
stimulated GLP-1 and PYY secretion in humans, &&
[12 ]. Reflex control of gastric emptying by CCK
but had no effect on CCK release [9]. Even so, the activation of vagovagal reflexes may, therefore,

1752-296X ß 2012 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-endocrinology.com 9

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Growth and development

reflect sequential paracrine and then hormonal leptin is reported to suppress fos labelling in PVN,
activation of different afferent pathways that are and also supra optic nucleus, in response to CCK
coupled to vagal efferent pathways controlling dis- (intraperitoneally), whereas there is no difference
tal and proximal gastric motility, respectively. in NTS labelling [20]; thus, although leptin and
Nodose ganglion neurons that express CCK-1 CCK may interact positively at the level of vagal
receptors also express several ion channels of the afferent neurons, there may also be other – possibly
transient receptor potential (TRP) class including direct – actions of leptin on hypothalamic neurons
TRPV1–5. Ruthenium red which is an inhibitor of that modulate the response to CCK.
this family of ion channels blocked CCK-induced
depolarization and increased intracellular calcium
in cultured rat nodose ganglion neurons, but a Cholecystokinin and vagal afferent plasticity
specific TRPV1 inhibitor (SB366791) had little effect, The vagal afferent neurons expressing CCK-1 recep-
suggesting that TRPV2–5 might be important in tors exhibit plasticity in their neurochemical pheno-
mediating the effects of CCK on nodose neurons type depending on nutrient status. Both acute and
[13]. Several actions of CCK on rat vagal afferent chronic changes have been described. In the rat,
neurons are potentiated by leptin, and it seems acute withdrawal of food over a period of 6 h or longer
possible that interactions between Src and PI-3 induces expression in nodose ganglion neurons of
kinase downstream of the CCK-1 receptor, and the neuropeptide transmitter melanin concentrating
STAT3 phosphorylation downstream of leptin hormone and of CB1 receptors (both associated
receptor targeted by leptin, act to regulate neuronal with orexigenic signalling), whereas there is reduced
excitation through closure of Kþ channels [14]. expression of the putative satiety neuropeptide
CARTp and of a population of Y2 receptors [3,21].
Both exogenous and endogenous CCK reversed these
Cholecystokinin actions on brain stem changes through mechanisms dependent on CCK-1
neurons receptor activation. Recent work suggests co-operat-
Recordings of miniature excitatory postsynaptic ive interactions between CCK and leptin to control
currents in neurons of the centralis division of the transmitter and receptor expression in these neurons.
rat NTS indicate a glutaminergic input from vagal Thus, expression of CART is mediated by a CCK–
afferent neurons. About 30% of neurons respond to leptin interaction involving the transcription factor
& &
CCK and all of these also respond to the vanilloid early growth response (EGR)-1 [22 ,23 ]. In unstimu-
agonist capsaicin. Both responses are substantially lated vagal afferent neurons, EGR-1 is localized to the
reduced by chemical lesioning of small diameter cytosol but in response to CCK there is nuclear trans-
afferent fibres using perivagal capsaicin [15] com- location which is strongly potentiated by leptin,
patible with work over many years, indicating that although leptin alone has no effect. Interestingly,
CCK inhibition of food intake and gastric emptying however, leptin strongly stimulates EGR-1 expres-
is mediated by capsaicin-sensitive vagal afferent sion, whereas CCK has little or no effect on EGR-1
neurons. Injection of N-methyl D-aspartate (NMDA) expression. It seems, then, that leptin determines the
receptor antagonists (MK-801, D-CPPene) into the magnitude of CCK stimulation of CART expression
fourth ventricle, or directly into the NTS, inhibited by controlling EGR-1 abundance. There remains
the effect of CCK on food intake, indicating a role some controversy over whether or not the gastric
for brainstem NMDA receptors in mediating the orexigenic hormone, ghrelin, inhibits CCK stimu-
responses to intraperitoneal CCK [16]. lation of vagal afferent discharge [24,25]; neverthe-
less, there is clear evidence that in rat nodose
ganglion neurons, ghrelin inhibits leptin stimulation
Hypothalamic responses to peripheral of EGR-1 expression at least in part by preventing
cholecystokinin STAT3 relocation to the nucleus and it also inhibits
Peripheral administration of CCK (intraperitoneally) CCK stimulation of CART expression by preventing
&
stimulates neurons in the paraventricular nucleus nuclear relocation of EGR-1 [23 ].
(PVN) of the hypothalamus. The PVN neurons acti-
vated by CCK express cocaine and amphetamine
regulated transcript (CART) and phosphor-mamma- CHOLECYSTOKININ AND GUT–BRAIN
lian target of rapamycin [17,18]. When given alone, SIGNALLING IN OBESITY
CCK does not increase fos labelling in the arcuate The satiety effect of CCK is attenuated in obesity; this
nucleus; however CARTp intracerebroventricularlly is accompanied by decreased activation of NTS
stimulates fos labelling in the arcuate and CCK (intra- neurons (compatible with decreased stimulation
peritoneally) augments this response [19]. Curiously, by vagal afferent neurons) [4]. There may also be

10 www.co-endocrinology.com Volume 19  Number 1  February 2012

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Cholecystokinin Dockray

attenuated splanchnic sympathetic nerve discharge there is decreased islet cell size and b-cell mass,
with implications for vasomotor control [26]. The suggesting a role for CCK in maintaining islet mass
mechanisms have been uncertain, but direct studies [31]. Interestingly, mice null for both glucagon
of vagal afferent neuron responses to CCK have now and GLP-1 receptors (Gcgr//Glp1r/) exhibit
shown in obese mice (high-fat diet for 8–10 weeks, increased expression of CCK-1 receptors in islets,
60% calories as fat) that there is reduced stimulation and increased sensitivity to CCK for insulin release
of jejunal afferent fibres by CCK and 5-hydroxytryp- [32]. Together, these studies indicate that CCK
tamine, reduced mechanosensitivity of low- either hormonally or as a paracrine/autocrine factor
threshold jejunal afferent fibres, reduced excitability might play a role in mediating compensatory mech-
of nodose ganglion neurons including those projec- anisms within the islets of Langerhans.
ting to the small intestine and decreased numbers of
nodose neurons showing increased intracellular
&&
calcium in response to CCK [27 ]. Insensitivity of CONCLUSION
vagal afferent neurons to CCK would, therefore, The release of CCK by intestinal I-cells in response to
appear to account for the reduced satiety effect of fatty acids and amino acids involves two G-protein-
CCK in obesity. Resistance to leptin also develops in coupled receptors: GPR40 and CaSR, respectively.
vagal afferent neurons in obesity, and interestingly Local release of CCK acts in a paracrine manner to
this occurs earlier than in CNS neurons [28]. The stimulate vagal afferent neurons, and subsequently
relationship between loss of responsiveness to CCK rising concentrations in the blood are associated with
and to leptin in obesity remains to be elucidated. hormonal stimulation of vagal afferent fibres to the
However, in CCK-1 receptor-null mice placed on a stomach. The stimulation of vagal afferent neurons is
high-fat diet, a short fast leads to decreased time to the associated with acute inhibition of food intake and
first meal on reintroduction of food and this increased gastric emptying and, over a period of a few hours,
drive to eat is mediated by ghrelin; moreover, a ghre- with changes in receptor and neuropeptide trans-
lin receptor antagonist, D-(Lys3)-growth hormone mitter expression in these neurons. Ghrelin inhibits,
releasing peptide-6 increased expression of CART in and leptin potentiates, many of these effects. The
vagal afferent neurons of CCK-1-null mice on a high- human CNS responses to ingestion of lipid are largely
fat diet (but not wild-type mice). The data suggest that mediated by release of CCK. In obesity, vagal afferent
the combination of a high-fat diet and loss of CCK-1 neurons exhibit resistance to the effect of CCK, appa-
receptors leads to increased ghrelin stimulation of rently due to decreased electrical excitability.
food intake in part by suppressing the expression of
a putative satiety peptide (CART) in vagal afferent
neurons [29]. Acknowledgements
Mice null for CCK appear to be resistant to diet- The author’s work in this area has been supported by
induced obesity. Thus, CCK/ mice on a high-fat grants from the Biotechnology and Biosciences Research
diet for 10 weeks exhibit reduced gain in body Council and the Wellcome Trust.
weight compared with wild type and reduced adi-
posity [30]. Although food intake was similar, the Conflicts of interests
phenotype may reflect lower energy extraction, as
There are no conflicts of interest.
there were defects in fat absorption. However,
energy expenditure was also higher in the CCK/
mice and there was greater oxidation of carbo- REFERENCES AND RECOMMENDED
hydrates on the high-fat diet, suggesting CCK is READING
involved in metabolic regulation, as well as lipid Papers of particular interest, published within the annual period of review, have
been highlighted as:
absorption, in response to high-fat diets. & of special interest
&& of outstanding interest

Additional references related to this topic can also be found in the Current
World Literature section in this issue (p. 68).
ENTEROINSULAR AXIS
1. Moran TH. Gut peptides in the control of food intake. Int J Obes (Lond) 2009;
The role of CCK as an insulin-releasing hormone, 33 (Suppl 1):S7–S10.
that is, incretin, has generally been considered to be 2. Raybould HE. Gut chemosensing: interactions between gut endocrine cells
and visceral afferents. Auton Neurosci 2010; 153:41–46.
of minor significance compared with other intesti- 3. Dockray GJ, Burdyga G. Plasticity in vagal afferent neurones during feeding
nal hormones such as GLP-1 and glucose-dependent and fasting: mechanisms and significance. Acta Physiol (Oxf) 2011;
201:313–321.
insulin releasing peptide. However, in the islets of 4. Covasa M. Deficits in gastrointestinal responses controlling food intake and body
obese mice (ob/ob on a C57Bl/6 background), there weight. Am J Physiol Regul Integr Comp Physiol 2010; 299:R1423–R1439.
5. de Lartigue G, Barbier de la Serre C, Raybould HE. Vagal afferent neurons in
is upregulation of CCK gene expression. Moreover, high fat diet-induced obesity; intestinal microflora, gut inflammation and
in CCK/ mice bred onto an ob/ob background, cholecystokinin. Physiol Behav 2011; 105:100–105.

1752-296X ß 2012 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-endocrinology.com 11

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Growth and development

6. Liou AP, Lu X, Sei Y, et al. The G-protein-coupled receptor GPR40 directly 19. Pirnik Z, Maixnerova J, Matyskova R, et al. Effect of anorexinergic peptides,
& mediates long-chain fatty acid-induced secretion of cholecystokinin. Gastro- cholecystokinin (CCK) and cocaine and amphetamine regulated transcript
enterology 2011; 140:903–912. (CART) peptide, on the activity of neurons in hypothalamic structures of
Long-chain fatty acids release CCK from intestinal I-cells. This article shows that C57Bl/6 mice involved in the food intake regulation. Peptides 2010; 31:139–
GPR40 is expressed by I-cells and acts as the sensor for fatty acid stimulation of 144.
CCK release. 20. Caquineau C, Douglas AJ, Leng G. Effects of cholecystokinin in the
7. Liou AP, Sei Y, Zhao X, et al. The extracellular calcium-sensing receptor is supraoptic nucleus and paraventricular nucleus are negatively modulated
required for cholecystokinin secretion in response to L-phenylalanine in by leptin in 24-h fasted lean male rats. J Neuroendocrinol 2010; 22:446–
acutely isolated intestinal I cells. Am J Physiol Gastrointest Liver Physiol 452.
2011; 300:G538–G546. 21. Burdyga G, Varro A, Dimaline R, et al. Expression of cannabinoid CB1
8. Wang Y, Chandra R, Samsa LA, et al. Amino acids stimulate cholecystokinin receptors by vagal afferent neurons: kinetics, and role in influencing neuro-
release through the Ca2þ-sensing receptor. Am J Physiol Gastrointest Liver chemical phenotype. Am J Physiol Gastrointest Liver Physiol 2010;
Physiol 2011; 300:G528–G537. 299:1514–1524.
9. Gerspach AC, Steinert RE, Schonenberger L, et al. The role of the gut sweet 22. de Lartigue G, Dimaline R, Varro A, et al. Cocaine and amphetamine regulated
taste receptor in regulating GLP-1, PYY and CCK release in humans. Am J & transcript mediates the actions of cholecystokinin on rat vagal afferent
Physiol Endocrinol Metab 2011; 301:E317–E325. neurons. Gastroenterology 2010; 138:1479–1490.
10. Little TJ, Gopinath A, Patel E, et al. Gastric emptying of hexose sugars: role of This article reports how CCK releases CART from vagal afferent neurons, and how
osmolality, molecular structure and the CCK receptor. Neurogastroenterol CART in turn enhances some of the effects of CCK on these neurons.
Motil 2010; 22:1183–1190; e314. 23. de Lartigue G, Lur G, Dimaline R, et al. EGR1 is a target for cooperative
11. Lassman DJ, McKie S, Gregory LJ, et al. Defining the role of cholecystokinin in & interactions between cholecystokinin and leptin, and inhibition by ghrelin, in
&& the lipid-induced human brain activation matrix. Gastroenterology 2010; vagal afferent neurons. Endocrinology 2010; 151:3589–3599.
138:1514–1524. The transcription factor EGR-1 is shown to be a target for co-operative interactions
This study used fMRI to define for the first time the human CNS responses to between CCK and leptin in vagal afferent neurons: leptin regulating EGR-1
ingestion of lipid. There is activation of the brain stem, hypothalamus and motor abundance and CCK its activation.
cortex that is strongly inhibited by a CCK receptor antagonist. 24. Date Y, Toshinai K, Koda S, et al. Peripheral interaction of ghrelin with
12. Okano-Matsumoto S, McRoberts JA, Tache Y, Adelson DW. Electrophy- cholecystokinin on feeding regulation. Endocrinology 2005; 146:3518–
&& siological evidence for distinct vagal pathways mediating CCK-evoked 3525.
motor effects in the proximal versus distal stomach. J Physiol 2011; 25. Arnold M, Mura A, Langhans W, Geary N. Gut vagal afferents are not
589:371–393. necessary for the eating-stimulatory effect of intraperitoneally injected ghrelin
By recording simultaneously and separately from vagal afferent and efferent in the rat. J Neurosci 2006; 26:11052–11060.
neurons serving different regions of the upper gastrointestinal tract, this study 26. How JM, Fam BC, Verberne AJ, Sartor DM. High-fat diet is associated with
has identified paracrine and endocrine actions of CCK on vagal afferent neurons, blunted splanchnic sympathoinhibitory responses to gastric leptin and cho-
and the different consequences for proximal and distal gastric motility. lecystokinin: implications for circulatory control. Am J Physiol Heart Circ
13. Zhao H, Simasko SM. Role of transient receptor potential channels in Physiol 2011; 300:H961–H967.
cholecystokinin-induced activation of cultured vagal afferent neurons. Endo- 27. Daly DM, Park SJ, Valinsky WC, Beyak MJ. Impaired intestinal afferent nerve
crinology 2010; 151:5237–5246. && satiety signalling and vagal afferent excitability in diet induced obesity in the
14. Heldsinger A, Grabauskas G, Song I, Owyang C. Synergistic interaction mouse. J Physiol 2011; 589:2857–2870.
between leptin and cholecystokinin in the rat nodose ganglia is mediated by There is resistance to the satiety effect of CCK in obesity and this study shows that
PI3K and STAT3 signaling pathways: implications for leptin as a regulator of it is attributable to decreased electrical excitability of vagal afferent neurons.
short term satiety. J Biol Chem 2011; 286:11707–11715. 28. de Lartigue G, Barbier de la Serre C, Espero E, Lee J, Raybould HE. Diet-
15. Browning KN, Wan S, Baptista V, Travagli RA. Vanilloid, purinergic and CCK induced obesity leads to the development of leptin resistance in vagal afferent
receptors activate glutamate release on single neurons of the nucleus tractus neurons. Am J Physiol Endocrinol Metab 2011; 301:E187–E195.
solitarius centralis. Am J Physiol Regul Integr Comp Physiol 2011; 29. Lee J, Martin E, Paulino G, et al. Effect of ghrelin receptor antagonist on meal
301:R394–R401. patterns in cholecystokinin type 1 receptor null mice. Physiol Behav 2011;
16. Wright J, Campos CA, Herzog T, et al. Reduction of food intake by 103:181–187.
cholecystokinin requires activation of hindbrain NMDA-type glutamate 30. Lo CM, King A, Samuelson LC, et al. Cholecystokinin knockout mice are
receptors. Am J Physiol Regul Integr Comp Physiol 2011; 301:R448– resistant to high-fat diet-induced obesity. Gastroenterology 2010;
R455. 138:1997–2005.
17. Peter L, Stengel A, Noetzel S, et al. Peripherally injected CCK-8S activates 31. Lavine JA, Raess PW, Stapleton DS, et al. Cholecystokinin is up-regulated in
CART positive neurons of the paraventricular nucleus in rats. Peptides 2010; obese mouse islets and expands beta-cell mass by increasing beta-cell
31:1118–1123. survival. Endocrinology 2010; 151:3577–3588.
18. Lembke V, Goebel M, Frommelt L, et al. Sulfated cholecystokinin-8 activates 32. Ali S, Lamont BJ, Charron MJ, Drucker DJ. Dual elimination of the glucagon
phospho-mTOR immunoreactive neurons of the paraventricular nucleus in and GLP-1 receptors in mice reveals plasticity in the incretin axis. J Clin Invest
rats. Peptides 2011; 32:65–70. 2011; 121:1917–1929.

12 www.co-endocrinology.com Volume 19  Number 1  February 2012

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

You might also like