You are on page 1of 5

Regulatory Peptides 155 (2009) 6–10

Contents lists available at ScienceDirect

Regulatory Peptides
j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / r e g p e p

Review

Cholecystokinin and gut–brain signalling


Graham J. Dockray ⁎
Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown St, Liverpool L69 3BX, UK

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

Article history: Enteroendocrine cells of the gastrointestinal tract act as a luminal surveillance system responding to either
Received 23 March 2009 the presence or absence of food in the gut lumen. Collectively, their secretory products regulate the course of
Accepted 25 March 2009 digestion and determine the delivery of nutrient to the gut by controlling food intake. Afferent neurons of the
Available online 2 April 2009
vagus nerve are an important target of gut hormones, particularly for control of food intake. The intestinal
hormone cholecystokinin (CCK) stimulates vagal afferent neuron discharge and also controls the expression
Keywords:
of both G-protein coupled receptors and peptide neurotransmitters in these neurons. When plasma CCK
Cholecystokinin
Vagus concentrations are low, for example in fasting, vagal afferent neurons express cannabinoid CB1 and melanin
Leptin concentrating hormone (MCH)-1 receptors, both of which are associated with stimulation of food intake.
PYY Post-prandial release of CCK rapidly down-regulates the expression of both receptors but stimulates the
Ghrelin expression of Y2 receptors in neurons projecting to the stomach. In fasting, there is also increased expression
Receptors in these neurons of the appetite-stimulating neuropeptide transmitter MCH, and depressed expression of the
Nodose ganglion satiety-peptide cocaine and amphetamine regulated transcript (CART). Secretion of CCK decreases
Satiety expression of MCH and increases expression of CART. The neurochemical phenotype of vagal afferent
Gastric emptying
neurons therefore encodes whether or not there has been nutrient ingestion over the previous period. At low
plasma concentrations of CCK vagal afferent neurons exhibit increased capacity for appetite-stimulation,
while post-prandial concentrations of CCK lead to enhanced capacity for satiety signalling. A gatekeeper
function can therefore be attributed to CCK in that its presence or absence influences the capacity of vagal
afferent neurons to respond to other neurohormonal signals.
© 2009 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
2. Integrative role of CCK in upper gastrointestinal tract physiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3. The neurochemical phenotype of vagal afferent neurons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4. The gatekeeper functions of CCK on vagal afferent neurons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
5. Modulating the gatekeeper function of CCK . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
6. Overview. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10

1. Introduction organised functional domains: the stomach, proximal small intestine


and distal small intestine/colon. In each case, the secretory products of
An impressive range of humoral factors signal changes in the luminal EECs ensure optimal digestion and absorption by controlling the nature
environment of the gastrointestinal tract [1]. The peptides and amines of of the luminal contents through stimulation or inhibition of digestive
the enteroendocrine cells (EECs) that act as primary transducers in secretions, and by regulating the delivery of nutrient along the
luminal surveillance are secreted in response to either the presence, or alimentary tract. In addition, it is now clear that there are many
absence, of luminal stimuli; they have been intensively studied for over a different immune modulators that respond to infection and inflamma-
century. Taken together they can be thought to function in three loosely tion in the gut wall and that act on some of the same targets as the
products of EEC secretion. Quite recently, a range of lipid mediators has
⁎ Tel.: +44 151 794 5324; fax: +44 151 1794 5315. emerged that also act as gut signals, these include the cannabinoid (CB)1
E-mail address: g.j.dockray@liverpool.ac.uk. agonist anandamide which stimulates food intake and the related

0167-0115/$ – see front matter © 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.regpep.2009.03.015
G.J. Dockray / Regulatory Peptides 155 (2009) 6–10 7

Fig. 1. Schematic representation of the neurohumoral factors acting on vagal afferent neurons. Leptin, CCK, PYY3-36 and GLP-1 are associated with stimulation of vagal afferent
pathways leading to inhibition of food intake and, or, gastric emptying. Ghrelin, orexin-A, anandamide and MCH are associated with stimulation of food intake and, or, gastric
emptying; CCK either inhibits expression of their receptors or they inhibit the actions of CCK.

oleylethanolamide which inhibits food intake [2,3]. Exactly how all of digestion of fat and protein in the small intestine by balancing the
these signalling molecules are integrated is an important issue for capacity to secrete enzyme and bile salt with the delivery of nutrient
present research. substrates. Although there are direct actions of CCK on pancreatic
Nutrient delivery to the small intestine is regulated by varying acinar cells and on gastric smooth muscle cells, it appears that afferent
the rate of gastric emptying and through control of food intake. In neurons of the vagus nerve are a target of CCK for reflex stimulation of
the latter case, there are inhibitors of food intake from each of the pancreatic secretion and inhibition of gastric emptying [13,14]. The
functional domains mentioned above (stomach: leptin; small same afferent pathway is thought to mediate the action of CCK in
intestine: CCK; ileum/colon: PYY, GLP-1) (Fig. 1) and stimulants inhibiting food intake [15]. Many studies have confirmed the
from two of them (stomach: ghrelin; small intestine: anandamide). observation of Moran et al. who demonstrated that vagal afferent
The two stimulants are released when the gut is empty and the neurons express CCK1 (or CCK-A) receptors and that these are
inhibitors are directly or indirectly released by luminal nutrients. In transported towards the periphery [16]. In addition to stimulation of
some circumstances gut hormones may act directly on CNS neurons vagal afferent nerve discharge CCK also controls the expression of both
after delivery in the circulation; however over the last decade or so, G-protein coupled receptors and peptide neurotransmitters involved
afferent neurons of the vagus nerve have emerged as important in controlling food intake thereby regulating the capacity of other
targets of gut hormones, particularly for control of food intake and neurohumoral agents to act on this pathway.
gastric emptying. There have been many recent reviews of the
literature on gut hormones and control of food intake [4–7]; the
present account will focus on the relationships between gut 3. The neurochemical phenotype of vagal afferent neurons
endocrine signals and vagal afferent neurons with a special emphasis
on CCK. A wide range of receptors and neuropeptides have been shown to
be expressed by vagal afferent neurons [6,17]. Work by Zhang et al.
established that the neurochemical phenotype of vagal afferent
2. Integrative role of CCK in upper gastrointestinal tract physiology neurons could be varied depending on experimental treatment.
They showed that vagotomy decreased CCK1 receptor expression
Ingestion of fat or protein rich meals leads to secretion of CCK from but increased expression of CCK2 (gastrin/CCK-B) and Y2 receptors
I-type EECs of the upper small intestine. Both in vivo and in cultured [18]; they also showed that vagotomy influenced the expression of
STC1 cells, fatty acids with a chain length greater than 12 carbons are genes encoding the regulatory peptides galanin, NPY, VIP and CCK
effective releasers of CCK while those with a chain length b10 C are itself, which normally exhibit low or moderate levels of expression
largely ineffective [8,9]. In human volunteers, a rise in plasma CCK [19,20]. Subsequently it has become clear that the neurochemical
concentrations of 3–4 pM can be achieved with modest loads of C12 phenotype of these neurons exhibits reversible changes in response to
and is sufficient to inhibit gastric emptying and decrease the intake of energy restriction.
a liquid test meal [10]. In addition to luminal nutrients, it is now clear In rats, withdrawal of food for periods of over 6 h has been shown
that there is also elevated plasma CCK in humans infected with small to produce changes in both receptor and neuropeptide gene
intestinal pathogens such as Giardia [11]. Moreover, in a mouse model expression in vagal afferent neurons [21–25]. It is possible to identify
of intestinal inflammation due to infection with the nematode Tri- three different patterns of gene expression following manipulation of
chinella spiralis, the secretion of CCK was shown to be dependent on energy intake. (a) There are some genes that exhibit broadly similar
CD4+ T-cells, via the release of IL-4 and IL-13 [12]. levels of expression in nodose ganglion neurons in rats that are fed ad
The primary actions of CCK are stimulation of pancreatic enzyme libitum or fasted up to 48 h (Fig. 2). For example, the expression of
secretion and gall bladder contraction, and inhibition of gastric CCK1, orexin type-1 (Ox-R1) and ghrelin receptors (GHS-1) seems not
emptying and food intake. Collectively, these actions allow optimal to be substantially altered with food withdrawal [20,21,26]. (b) Some
8 G.J. Dockray / Regulatory Peptides 155 (2009) 6–10

Fig. 2. Cannabinoid (CB)-1 receptor expression in vagal afferent neurons is increased by energy restriction. A, In situ hybridisation demonstrates expression of CB1 mRNA in neurons
of the nodose ganglion in fasted rats, but not rats fed ad libitum (control). In contrast CCK-1 receptor expression is similar in fasted and fed ad libitum rats. B, Retrograde tracing from
the stomach with True Blue, coupled with double labelling immunohistochemistry indicates that nodose ganglion neurons projecting to the stomach express both CB1 and CCK-1
receptors. Reproduced from Journal of Neuroscience.

genes exhibit increased expression in nodose ganglion neurons in rats expression dependent on the presence or absence of CCK. Administra-
following food withdrawal; examples of this class include the tion of CCK to fasted rats stimulates expression of CART and Y2
cannabinoid (CB)1 and melanin concentrating hormone (MCH)1 receptors, and inhibits expression of CB1, MCH1 and MCH [21–23,25].
receptors and MCH itself; each of these is associated with orexigenic The effects seen with refeeding of fasted rats on expression of CB1,
signalling in the CNS. (c) A third group exhibits depressed expression MCH1 and Y2 receptors, and on the neuropeptides MCH and CART, are
in fasted rats and is increased by refeeding eg cocaine and attributable to the action of endogenous CCK since in each case they are
amphetamine regulated transcript (CART) and Y2 receptors both of blocked by the administration of a CCK1 receptor antagonist [21–23].
which are associated with satiety signalling [21–23,26]. Refeeding of In cultured vagal afferent neurons, the neurochemical plasticity
fasted rats reverses the neurochemical phenotype relatively rapidly seen in vivo can be replicated by culturing in media either with or
[21–23,25,26]. The neurochemical phenotype of vagal afferent without fetal calf serum (FCS). For example, in full media (10% FCS)
neurons exhibits, therefore, a simple form of memory by encoding there is expression of CART but not MCH, while in serum-free medium
whether or not there has been nutrient ingestion over the previous there is expression of MCH but not CART [23]. Addition of CCK to
period (Fig. 3). neurons in serum-free medium rapidly restores CART expression and
decreases MCH expression. The action of CCK in stimulating CART
4. The gatekeeper functions of CCK on vagal afferent neurons depends on activation of PKC, phosphorylation of CREB and activation
of MAPkinase. The mechanisms of down-regulation are less clear, but
It is well established that CCK stimulates the discharge of vagal neurons expressing a CREB inhibitor, seem not to demonstrate down-
afferent neurons and increases intracellular calcium concentrations regulation of MCH suggesting that CREB is on both the stimulatory and
[27–29]. In addition, though, recent work has identified changes in gene inhibitor pathways downstream of CCK [23].
G.J. Dockray / Regulatory Peptides 155 (2009) 6–10 9

Fig. 3. The neurochemical phenotype of vagal afferent neurons depends on energy intake. In rats fed ad libitum (right) there is expression of Y2 receptors and the satiety-peptide
CART in vagal afferent neurons projecting to the stomach, but not MCH-1 and CB1 receptors or MCH. After energy restriction for 6–48 h there is progressive up regulation of MCH-1
and CB1 receptors, and MCH, with loss of Y2 receptors and CART; these changes are reversed by refeeding or administration of CCK. The abundance of CCK-1, Ob-r, GHS-1 and Ox-R1
receptors seems to be similar in fed and fasted rats.

The actions of CCK in controlling Y2 receptor expression are so far as its presence or absence influences the capacity for responding to
interesting. When rats are fasted there is a progressive loss of other neurohormonal signals.
expression of Y2 receptors with a t1/2 of about 12 h. However, even
after 48 h of food withdrawal there is a small population of neurons in 5. Modulating the gatekeeper function of CCK
which expression is preserved that amounts to 5–10% of the total
population in the mid and caudal regions of the nodose ganglion. The action of CCK in stimulating vagal afferent neuron discharge
Studies combining retrograding tracing with immunohistochemistry and inhibiting food intake is potentiated by gastric distension, leptin
have shown that the neurons exhibiting loss of Y2 receptors with food and urocortin [28,29,31–34]. In contrast, orexin-A and ghrelin inhibit
restriction mostly project to the stomach, whereas the vagal afferent the action of CCK on these neurons [35,36]. During the immediate
neurons serving the ileum and colon continue to express these post-prandial period when CCK concentrations are rising and ghrelin
receptors during food withdrawal. The endogenous ligand of Y2 concentrations are falling, vagal afferent neurons are exposed to both
receptors is PYY3-36 produced by secretion of PYY from EECs and interactions between the two are therefore possible. Although in
predominantly located in the ileum and colon. The vagal afferent principle the peptide obestatin derived from the ghrelin precursor is
neurons projecting to these regions are therefore expected to be also likely to be secreted alongside ghrelin, recent work suggests it has
exposed to locally high concentrations of PYY3-36, and receptor no effect on CCK-satiety signalling [37]. The action of CCK in down-
activation is not dependent on CCK, while those to the stomach regulating CB1, MCH1 and MCH is inhibited by prior administration of
presumably respond to PYY3-36 delivered as a hormone in the ghrelin [26]; similarly, CCK-stimulated increases in CART and Y2
circulation and signalling is subject to prior stimulation of receptor receptor expression are blocked by ghrelin administration [23]. In
expression by CCK. It is worth noting that the action of PYY3-36 both cultured vagal afferent neurons, ghrelin has been shown to act by
in delaying gastric emptying and in stimulating fos expression in brain excluding phosphoCREB from the nucleus [23]. The cellular mechan-
stem neurons (which is a putative marker of vagal afferent excitation) isms are unknown but merit further study.
is CCK-dependent [25,30]; these observations are therefore consistent
with the role of CCK in controlling Y2 receptor expression in vagal 6. Overview
afferent neurons serving the stomach.
Together, these data indicate that CCK is able to switch the Vagal afferent neurons seem able to adopt two states: one
neurochemical phenotype of vagal afferent neurons between two states. associated with expression of signalling molecules associated with
At low plasma concentrations (through energy restriction), the vagal inhibition of food intake, the other with signalling molecules
afferent pathway exhibits increased capacity for appetite-stimulation, associated with stimulation of food intake. These states are deter-
while post-prandial concentrations of CCK lead to enhanced capacity for mined by energy intake over the previous day or so. In addition to the
satiety signalling. Thus a gatekeeper function can be ascribed to CCK in acute actions of CCK on vagal afferent neurons, it seems that there are
10 G.J. Dockray / Regulatory Peptides 155 (2009) 6–10

also longer term effects over hours or even days in which CCK switches [19] Zhang X, Ji RR, Arvidsson J, Lundberg JM, Bartfai T, Bedecs K, Hokfelt T. Expression
of peptides, nitric oxide synthase and NPY receptor in trigeminal and nodose
neurons between the two states. Manipulation of these states could ganglia after nerve lesions. Exp Brain Res 1996;111:393–404.
provide a novel therapeutic target for the control of food intake and [20] Broberger C, Holmberg K, Shi TJ, Dockray G, Hokfelt T. Expression and regulation of
the treatment of obesity. cholecystokinin and cholecystokinin receptors in rat nodose and dorsal root
ganglia. Brain Res 2001;903:128–40.
[21] Burdyga G, Lal S, Varro A, Dimaline R, Thompson DG, Dockray GJ. Expression of
References cannabinoid CB1 receptors by vagal afferent neurons is inhibited by cholecysto-
kinin. J Neurosci 2004;24:2708–15.
[1] Dockray GJ. Gastrointestinal hormones: gastrin, cholecystokinin, somatostatin and [22] Burdyga G, Varro A, Dimaline R, Thompson DG, Dockray GJ. Feeding-dependent
ghrelin. In: Johnson LR, editor. Physiology of the gastrointestinal tract. Amsterdam: depression of melanin-concentrating hormone and melanin-concentrating hor-
Elsevier Academic Press; 2006. p. 91–120. mone receptor-1 expression in vagal afferent neurones. Neuroscience
[2] Gomez R, Navarro M, Ferrer B, Trigo JM, Bilbao A, Del AI, Cippitelli A, Nava F, 2006;137:1405–15.
Piomelli D, Rodriguez dF. A peripheral mechanism for CB1 cannabinoid receptor- [23] de Lartigue G, Dimaline R, Varro A, Dockray GJ. Cocaine- and amphetamine-
dependent modulation of feeding. J Neurosci 2002;22:9612–7. regulated transcript: stimulation of expression in rat vagal afferent neurons by
[3] Rodriguez dF, Navarro M, Gomez R, Escuredo L, Nava F, Fu J, Murillo-Rodriguez E, cholecystokinin and suppression by ghrelin. J Neurosci 2007;27:2876–82.
Giuffrida A, LoVerme J, Gaetani S, Kathuria S, Gall C, Piomelli D. An anorexic lipid [24] Buyse M, Ovesjo ML, Goiot H, Guilmeau S, Peranzi G, Moizo L, Walker F, Lewin MJ,
mediator regulated by feeding. Nature 2001;414:209–12. Meister B, Bado A. Expression and regulation of leptin receptor proteins in afferent
[4] Dockray GJ. Gut endocrine secretions and their relevance to satiety. Curr Opin and efferent neurons of the vagus nerve. Eur J Neurosci 2001;14:64–72.
Pharmacol 2004;4:557–60. [25] Burdyga G, de Lartigue G, Raybould HE, Morris R, Dimaline R, Varro A, Thompson DG,
[5] Murphy KG, Bloom SR. Gut hormones and the regulation of energy homeostasis. Dockray GJ. Cholecystokinin regulates expression of Y2 receptors in vagal afferent
Nature 2006;444:854–9. neurons serving the stomach. J Neurosci 2008;28:11583–92.
[6] Raybould HE. Mechanisms of CCK signaling from gut to brain. Curr Opin Pharmacol [26] Burdyga G, Varro A, Dimaline R, Thompson DG, Dockray GJ. Ghrelin receptors in rat
2007;7:570–4. and human nodose ganglia: putative role in regulating CB-1 and MCH receptor
[7] Chaudhri OB, Salem V, Murphy KG, Bloom SR. Gastrointestinal satiety signals. abundance. Am J Physiol Gastrointest Liver Physiol 2006;290:G1289–97.
Annu Rev Physiol 2008;70:239–55. [27] Blackshaw LA, Grundy D. Effects of cholecystokinin (CCK-8) on two classes of
[8] McLaughlin J, Grazia LM, Jones MN, D'Amato M, Dockray GJ, Thompson DG. Fatty gastroduodenal vagal afferent fibre. J Auton Nerv Syst 1990;31:191–201.
acid chain length determines cholecystokinin secretion and effect on human [28] Schwartz GJ, McHugh PR, Moran TH. Integration of vagal afferent responses to
gastric motility. Gastroenterology 1999;116:46–53. gastric loads and cholecystokinin in rats. Am J Physiol 1991;261:R64–9.
[9] McLaughlin JT, Lomax RB, Hall L, Dockray GJ, Thompson DG, Warhurst G. Fatty [29] Peters JH, Karpiel AB, Ritter RC, Simasko SM. Cooperative activation of cultured vagal
acids stimulate cholecystokinin secretion via an acyl chain length-specific, Ca2+- afferent neurons by leptin and cholecystokinin. Endocrinology 2004;145:3652–7.
dependent mechanism in the enteroendocrine cell line STC-1. J Physiol [30] Whited KL, Tso P, Raybould HE. Involvement of apolipoprotein A-IV and
1998;513:11–8. cholecystokinin1 receptors in exogenous peptide YY3 36-induced stimulation of
[10] Lal S, McLaughlin J, Barlow J, D'Amato M, Giacovelli G, Varro A, Dockray GJ, intestinal feedback. Endocrinology 2007;148:4695–703.
Thompson DG. Cholecystokinin pathways modulate sensations induced by gastric [31] Schwartz GJ, McHugh PR, Moran TH. Gastric loads and cholecystokinin synergistically
distension in man. Am J Physiol Gastrointest Liver Physiol 2004;287:G72–9. stimulate rat gastric vagal afferents. Am J Physiol 1993;265:R872–6.
[11] Leslie FC, Thompson DG, McLaughlin JT, Varro A, Dockray GJ, Mandal BK. Plasma [32] Gourcerol G, Wang L, Wang YH, Million M, Tache Y. Urocortins and cholecysto-
cholecystokinin concentrations are elevated in acute upper gastrointestinal kinin-8 act synergistically to increase satiation in lean but not obese mice:
infections. Q J Med 2003;96:870–1. involvement of corticotropin-releasing factor receptor-2 pathway. Endocrinology
[12] McDermott JR, Leslie FC, D'Amato M, Thompson DG, Grencis RK, McLaughlin JT. 2007;148:6115–23.
Immune control of food intake: enteroendocrine cells are regulated by CD4+ T [33] Wang YH, Tache Y, Sheibel AB, Go VL, Wei JY. Two types of leptin-responsive gastric
lymphocytes during small intestinal inflammation. Gut 2006;55:492–7. vagal afferent terminals: an in vitro single-unit study in rats. Am J Physiol
[13] Li Y, Owyang C. Endogenous cholecystokinin stimulates pancreatic enzyme 1997;273:R833–7.
secretion via vagal afferent pathway in rats. Gastroenterology 1994;107:525–31. [34] Barrachina MD, Martinez V, Wang L, Wei JY, Tache Y. Synergistic interaction
[14] Forster ER, Green T, Elliot M, Bremner A, Dockray GJ. Gastric emptying in rats: role between leptin and cholecystokinin to reduce short-term food intake in lean mice.
of afferent neurons and cholecystokinin. Am J Physiol 1990;258:G552–6. Proc Natl Acad Sci U S A 1997;94:10455–60.
[15] Smith GP, Jerome C, Cushin BJ, Eterno R, Simansky KJ. Abdominal vagotomy blocks [35] Burdyga G, Lal S, Spiller D, Jiang W, Thompson DG, Attwood S, Saeed S, Grundy D,
the satiety effect of cholecystokinin in the rat. Science 1981;213:1036–7. Varro A, Dimaline R, Dockray GJ. Localization of orexin-1 receptors to vagal
[16] Moran TH, Smith GP, Hostetler AM, McHugh PR. Transport of cholecystokinin (CCK) afferent neurons in the rat and humans. Gastroenterology 2003;124:129–39.
binding sites in subdiaphragmatic vagal branches. Brain Res 1987;415:149–52. [36] Date Y, Toshinai K, Koda S, Miyazato M, Shimbara T, Tsuruta T, Niijima A, Kangawa K,
[17] Dockray GJ. The brain–gut axis. In: Yamada T, editor. Textbook of Gastroenterology. Nakazato M. Peripheral interaction of ghrelin with cholecystokinin on feeding
New York: Raven Press; 2006. p. 77–92. regulation. Endocrinology 2005;146:3518–25.
[18] Zhang X, Shi T, Holmberg K, Landry M, Huang W, Xiao H, Ju G, Hokfelt T. Expression [37] Gourcerol G, Million M, Adelson DW, Wang Y, Wang L, Rivier J, St-Pierre DH, Tache
and regulation of the neuropeptide Y Y2 receptor in sensory and autonomic Y. Lack of interaction between peripheral injection of CCK and obestatin in the
ganglia. Proc Natl Acad Sci U S A 1997;94:729–34. regulation of gastric satiety signaling in rodents. Peptides 2006;27:2811–9.

You might also like