Vous êtes sur la page 1sur 12

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/287810606

Enteroendocrine Cells: A Review of Their Role In Brain-Gut Communication

Article  in  Neurogastroenterology and Motility · December 2015


DOI: 10.1111/nmo.12754

CITATIONS READS

39 303

4 authors, including:

Rocco Latorre Catia Sternini


Columbia University University of California, Los Angeles
42 PUBLICATIONS   362 CITATIONS    217 PUBLICATIONS   6,957 CITATIONS   

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Rocco Latorre on 13 June 2016.

The user has requested enhancement of the downloaded file.


Neurogastroenterology & Motility
Neurogastroenterol Motil (2015) doi: 10.1111/nmo.12754

REVIEW ARTICLE

Enteroendocrine cells: a review of their role in brain–


gut communication

R. LATORRE ,* C. STERNINI ,† R. DE GIORGIO ‡ & B. GREENWOOD-VAN MEERVELD *,§

*Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
†CURE Digestive Diseases Research Center, Division of Digestive Diseases and Departments of Medicine and Neurobiology,
David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
‡Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
§Department of Physiology, Veterans Affairs Medical Center, University of Oklahoma Health Sciences Center, Oklahoma City,
OK, USA

Key Messages

• Enteroendocrine cells (EECs) located in the entire GI tract are involved in luminal chemosensing and new
evidence suggests that they have a role in sensory nerve sensitization.
• EECs have direct cell connections to the nervous system. The EEC basal cytoplasmic processes interact with
the enteric nervous system (ENS), vagus nerve, and spinal afferent fibers, supporting their participation in the
complex and sophisticated activation of brain–gut axis bidirectional pathways.
• Perturbation of this critical communication between EECs, ENS and central nervous system might alter the
primary physiological and homeostatic gut functions.

Abstract enteric and extrinsic neurons. Recent studies have


Background Specialized endoderm-derived epithelial shed light on EEC sensory transmission by showing
cells, that is, enteroendocrine cells (EECs), are widely direct connections between EECs and the nervous
distributed throughout the gastrointestinal (GI) tract. system via axon-like processes that form a well-
Enteroendocrine cells form the largest endocrine organ defined neuroepithelial circuits through which EECs
in the body and play a key role in the control of GI can directly communicate with the neurons innervat-
secretion and motility, the regulation of food intake, ing the GI tract to initiate appropriate functional
postprandial glucose levels and metabolism. EECs responses. Purpose This review will highlight the role
sense luminal content and release signaling molecules played by the EECs in the complex and integrated
that can enter the circulation to act as classic sensory information responses, and discuss the new
hormones on distant targets, act locally on neighbor- findings regarding EECs in the brain–gut axis bidirec-
ing cells and on distinct neuronal pathways including tional communication.

Keywords afferent neurons, gut chemosensing,


Address for Correspondence peptides.
Rocco Latorre, PhD, Oklahoma Center for Neuroscience,
University of Oklahoma Health Science Center, 975 N.E. 10th Abbreviations: 5-HT, 5-hydroxytryptamine/serotonin;
Street, BRC 272, Oklahoma City, OK 73104, USA. CaSR, calcium sensing receptor; CCK, cholecys-
Tel.: +1 (405) 456 3547; Fax: 405 290 1719; tokinin; CNS, central nervous system; EC, enterochro-
e-mail: rocco-latorre@ouhsc.edu
maffin cell; EECs, enteroendocrine cells; FFARs, free
Received: 6 August 2015
Accepted for publication: 17 November 2015 fatty acid receptors; GFP, green fluorescent protein;

© 2015 John Wiley & Sons Ltd 1


R. Latorre et al. Neurogastroenterology and Motility

GI, gastrointestinal tract; GIP, gastric inhibitory pep- EECS: MORPHOLOGY, LOCALIZATION
tide; GLP-1, glucagon-like peptide-1; GLP-1R, gluca- AND FUNCTION
gon-like peptide-1 receptor; GLP-2, glucagon-like
Enteroendocrine cells are specialized endoderm-derived
petide-2; GPCRs, G protein-coupled receptor; NPY,
epithelial cells that differentiate from pluripotent stem
neuropeptide Y; NTS, nucleus of the solitary tract;
cells at the base of intestinal crypts and migrate up the
PYY, peptide YY; SCFA, short-chain fatty acid.
crypt-villus axis.15 The wide distribution of EECs in the
villi and crypts, where they are interspersed between
non-endocrine cells, is regulated by an interplay of
INTRODUCTION different factors including the cell surface protein
The endocrine system is distributed throughout the ‘notch’ and the basic helix-loop-helix transcription
human body, providing a constant control of physiolog- factor family, which control terminal cell differentia-
ical and homeostatic functions by secreting hormones, tion.15,16 Depending on their individual morphology and
which act on different cell targets. The largest endocrine position in the GI mucosa, EECs are divided into open
system in terms of number of cells is located in the type with a bottle neck shape and an apical prolongation
gastrointestinal (GI) tract, where it is comprised of with microvilli facing toward the intestinal lumen or
individually scattered enteroendocrine cells (EECs) dis- closed type that are located close to the basal membrane,
tributed along the entire GI mucosa in the crypts and do not reach the lumen of the gut and lack micro-
villi representing 1% of the total gut epithelium cell villi.3,17,18 The open type EECs directly detect luminal
population.1–3 Enteroendocrine cells are specialized contents through the microvilli reaching the lumen,
cells capable of sensing luminal content, producing whereas the close types are believed to be activated by
and releasing hormones/signaling molecules, and mod- luminal content indirectly either through neural or
ulating a variety of physiological GI and homeostatic humoral pathways. Both open- and closed-type cells
functions. Despite their well-established role, the accumulate their secretory products in cytoplasmatic
molecular sensing mechanisms and the modalities of granules and release them by exocytosis at the basolat-
transmission are not completely understood. However, eral membrane upon mechanical, chemical, or neural
there is increasing evidence that EECs are equipped with stimulation. The secretory products, mainly peptides/
a chemosensory machinery to initiate appropriate func- hormones, can act locally in a paracrine manner,
tional responses through gut–brain and brain–gut inter- activating other EECs and other cell types in the
action and that transmission involves at least in part mucosa, reach distant targets through release in the
direct EEC-neurons connections via basal cytoplasmic bloodstream or act directly on nerve endings close to the
processes. The first report of EEC basal processes was in site of release.11,17 Evidence for direct EEC-nerve trans-
1979 when Larsson et al. described basal cytoplasmic mission has been provided by Boh orquez et al.19,20 who
prolongation in somatostatin-containing EEC that have combined high-resolution imaging with transgenic
appeared to contact other cells thus providing evidence mice with EECs expressing green fluorescent protein
for a paracrine function of somatostatin D cells.4 Similar (GFP) and shown that basal processes of I and L cells have
processes of various lengths have been reported by other synaptic features, are accompanied by glia cells and
investigators in L cells containing polypeptide YY (PYY) contact nerves innervating the mucosa forming neu-
in addition to somatostatin D cells.5–7 Although basal roepithelial circuits.
processes are prominent in L and D cells, in other types Distinct types of EECs expressing more than 20
of EECs such as I cells secreting cholecystokinin (CCK) peptides/hormones have been identified along the GI
basal processes are typically short.8 Independently of the mucosa (Fig. 1) and it is now clear that many EECs
prominence of the basal processes, direct or indirect contain more than one signaling molecule.1,10,11 Evi-
communication of EECs with nerves is a major mech- dence for a complex phenotype of EECs is supported by
anism underlying EEC function in the gut mucosa studies using transgenic CCK-eGFP mouse, which
through peptide release.9–11 showed the broad co-expression of six functionally
Recently, there has been considerable interest in the different peptides (CCK, gastric inhibitory peptide
role of EECs in gut–brain/brain–gut communication [GIP], secretin, glucagon-like peptide-1 [GLP-1],
and the reader is referred to a series of excellent review PYY and neurotensin) in intestinal mucosa EECs.21
articles.12–14 The present review focuses on EECs Other studies combining transcriptional profiles,
function in nutrient sensing and food intake regulation fluorescence-activated cell sorting analysis and cell
emphasizing their role in brain–gut bidirectional culture systems have shown that cells expressing the
communication. same peptide/hormone vary depending on the region of

2 © 2015 John Wiley & Sons Ltd


EEC cells in the brain-gut communication

the intestine where they are located.22 The complexity this review, we have elected to use the classic
of EEC phenotypes has been further confirmed by a nomenclature based on the major hormone secreted
study using triple labeling immunohistochemistry by the different EEC subtypes throughout the gut
which has reported that in both mouse and pig small mucosa.
intestine and colon, a subset of L and K cells overlaps Enteroendocrine cell secretory products are released
(hence also referred to as K/L or L/K EECs) with in response to diverse types of stimuli and influence
different gradients of hormonal marker co-localization a variety of physiological functions. For instance,
(i.e., GLP-1, PYY and GIP).23 These findings clearly gastrin, secreted by open-type G cells of the gastric
indicate that EEC type classification based on the antrum and pylorus in response to luminal aminoacids
content of the cytoplasmic granules/vesicles is flawed and calcium, controls gastric acid secretion by acting
and that a nomenclature review is necessary. However, on closed-type enterochromaffin-like cells of the
the factors that determine how the EECs differentia- gastric corpus, which release histamine. In turn,
tion process occurs are still largely unknown and there histamine activates parietal cells to secret gastric
is also evidence of plasticity of the EEC system.18 In acid.24 Somatostatin, which is released by closed-type

Figure 1 Enteroendocrine cells (EECs) Subpopulations in the Gastrointestinal Tract. EECs comprise different subgroups producing and releasing a
variety of hormones under appropriate stimulation. EECs are largely distributed in the gut. The stomach corpus is rich in A cells secreting ghrelin
(dark red circle), the antrum is populated by G cells secreting gastrin (purple circle), while in the pylorum there is a high density of D cells secreting
somatostatin (red circle). D cells are also present in the proximal segment of the small intestine. I (dark blue circle) and K (orange circle) cells, which
secrete CCK and GIP, respectively, are located in the small intestine and L cells secreting GLP-1, GLP-2 and PYY (light blue circle) are widely
distributed in the distal small intestine and in the colon (mainly in the proximal portion). Up to 95% of the total 5-HT is produced in the GI tract. EC
(green circle) are situated in the pylorus, small and large intestine. 5-HT is also released by I cells (dark blue circle) located in the distal portion of the
small intestine and L cells (light blue circle) in the large intestine. Only some example of EECs subgroups and relative hormones secreted are
represented in the figure. Abbreviations: EC, enterochromaffin cell; 5-HT, 5-hydroxytryptamine; CCK, cholecystokinin; GLP-1, glucagon like
peptide-1; GLP-2, Glucagon like peptide-2; PYY, peptide YY; GIP, gastric inhibitory peptide.

Rapresentation: Open type EEC

Closed-type EEC

Open type EEC with short or long basal cytoplasmic processes.

© 2015 John Wiley & Sons Ltd 3


R. Latorre et al. Neurogastroenterology and Motility

D cells of the gastric corpus in response to intestinal in mind that, as stated above, subsets of EECs overlap
hormones such as CCK or transmitters, inhibits gastric in term of their secretory products supporting that
acid secretion by direct inhibition on parietal cells and different peptides may affect each other function once
through inhibition of histamine release.24 By contrast, released in response to luminal changes.22,23
somatostatin released from open type D cells in the
antrum inhibits gastric acid secretion through
EECS AS KEY REGULTORS OF GUT
inhibition of the production and release of gastrin.25
CHEMOSENSING, FOOD INTAKE, AND
5-hydroxytryptamine/serotonin (5-HT) is a polyfunc-
ENERGY HOMEOSTASIS
tional signaling molecule contained in the closed-type
enterochromaffin cells (EC) and in a subgroup of open Enteroendocrine cells respond to a wide range of
type I and L cells and in enteric neurons, mostly substances present in the lumen, classifiable as nutri-
interneurons.26,27 5-HT has been regarded as playing a ents, non-nutrient chemicals, food-born toxins, and
major role in promoting intestinal motility through a microorganisms.10,44 Enteroendocrine cells represent
combination of neuronal and mucosal mechanisms. the first level of integration of the information from the
The classic notion that 5-HT released from ECs upon gut lumen and upon stimulation release signaling
chemical stimulation or mechanical distortion initi- molecules that activate neuronal circuits, which send
ated the peristaltic reflex and propulsion has been information to different regions of the brain to generate
challenged.26–29 Investigators have suggested that 5- appropriate functional responses.1–3 Chemosensing of
HT-neurons play a more important role in gut motility luminal contents elicits a variety of functions, includ-
than 5-HT-EC cells though other studies are in ing digestion and absorption of nutrients and defense
disagreement with this conclusion.27 However, other responses against harmful/toxic substances such as
functions have been recently ascribed to mucosal (EC- vomiting, diarrhea, and food aversion. A major function
derived) 5-HT, including worsening inflammation in a of EEC-secreted peptides/hormones is to coordinate the
rat model of experimental colitis, promoting hepatic response of the gut to ingested nutrients. This includes
regeneration, lowering bone mass by inhibition of induction of GI, pancreatic and biliary secretion, and
osteoblast proliferation and serving as a growth fac- modulation of GI motility to facilitate digestion and
tor.27,30 The satiety hormone, leptin secreted by gastric absorption and other physiological activities such as
chief cells and open-type P cells,31 and the orexigenic tissue growth and repair, and increase in the intestinal
hormone, ghrelin secreted by closed- and open-type X/ barrier function through the activation of local and
A-like cells, regulate appetite.31,32 Ghrelin cells are neuronal pathways and the brain–gut axis.3,10,11 By
mostly abundant in the stomach, where they have the contrast, potentially harmful substances in the lumen
morphology of the closed-type, whereas in the small are likely to initiate a protective response possibly via
and large intestine there are both types with an EEC activation to reduce, reject, or avoid the threat by
increasing gradient of open-type cells toward the large delaying gastric emptying, increasing intestinal secre-
intestine, though the physiological significance of this tion, inducing vomiting, diarrhea, or food aversion
finding is unknown.33 Glucagon-like peptide-1, GLP-2 presumably through the activation of vagal afferents
and PYY, are contained in open type L cells and are and neurons of the caudal (visceral) nucleus of the
released in response to ingested nutrients, including solitary tract (NTS), parabrachial nucleus, central
carbohydrates and fat. Glucagon-like peptide-1 regu- nucleus of the amygdala, and paraventricular nucleus
lates postprandial glucose levels, promotes satiety and of the hypothalamus as shown by intraluminal admin-
has an inhibitory effect on energy intake.34 Glucagon- istration of bitter tastants.45–48 For instance, rotavirus
like peptide-2 regulates intestinal lipid absorption, encoded enterotoxin, a major cause of gastroenteritis in
promotes epithelial growth and exerts mucosal children, is likely to cause emesis, a hallmark of the
defense.35–37 Peptide YY3-36 inhibits food intake and virus-induced illness, by acting on EC cells with
induces food aversion,38,39 and in combination with subsequent release of 5-HT, which activates areas of
GLP-1 plays a major role in the ileal brake, a physio- the NTS and area postrema in the brain stem associated
logical mechanism delaying gut motility and transit in with nausea and vomiting via vagal activation.49
order to allow for nutrient (mainly lipids) absorp-
tion.32,40–42 Gastric inhibitory peptide secreted by open
Sensory receptors expressed on EECs
type K cells and by L cells containing GLP-1 and/or
PYY leads to the release of insulin in the postprandial Enteroendocrine cells have a key role in gut chemosens-
phase of digestion thus participating in the postpran- ing and are equipped with a wide array of receptors
dial glucose level control.32,40,43 It is important to keep expressed on the luminal side of the gut mucosa, many

4 © 2015 John Wiley & Sons Ltd


EEC cells in the brain-gut communication

of which are G protein-coupled receptors (GPCRs) and acids and are associated with different types of EECs,
respond to selective luminal substances.10,11 such as X/A, P, I, K, and L cells and EC-containing 5-
HT.62–64 In the colon, non-digestible carbohydrates are
processed by bacterial fermentation with production of
Taste receptors
short-chain fatty acids (SCFA) detected by free fatty
Enteroendocrine cell sensory receptors include taste acid receptors (FFARs).65 Activation of FFAR1, 2, 3
receptors and taste signaling molecules implicated in (also known as GPR40, 43 and 41, respectively), and
taste transduction originally described in taste buds of CaSR induce release of CCK to increase satiety and
the mouth where initial gustatory processing delay gastric emptying. Glucagon-like peptide-1 and
occurs.3,50–54 These include two major families of taste PYY are also secreted in response to FFAR1,2,3,
detecting GPCRs, the T1Rs sensing sweet and umami, activation.66–69 Thus, nutrients and non-nutrient
and the T2Rs detecting bitter taste.3,52,55 The T1R tastants are detected by EECs in the entire GI system
family is composed of three isoforms that function as from the proximal stomach to the rectum. In addition,
heterodimers, the T1R1-T1R3 dimer detects umami recent data indicate that also the ‘gut microbiota’, that
taste, while the T1R2-T1R3 dimer senses sweet taste, is, the myriad of bacteria – and viruses and fungi –
including glucose and artificial sweeteners.53,55,56 Gut present in diverse abundance throughout the gut
T1Rs recognize ingested nutrients such as carbohy- lumen, contribute to gut chemosensitivity.70,71 Indeed,
drates (T1R2-T1R3) and amino acids (T1R1-T1R3). bacterial products including SCFA metabolites act as a
Studies indicate that the gut T1R2-T1R3 may control ligand and activate specific receptors such as GPR41
glucose homeostasis through a combination of GLP-1 and GPR43 located on EECs, triggering the release of
release and glucose transporter activation,53 however, GI hormones (e.g., GLP-1, GLP-2, and PYY) and a
there is still controversy about the exact role of T1Rs subsequent regulation of physiological, homeostatic,
in glucose homeostasis.11 T1R1-T1R3 have been impli- and barrier functions.70,72 Bacterial metabolites are
cated in appetite/satiety regulation by releasing CCK also recognized by toll-like receptors, some of which
through a pathway that might involve the gut–brain have been localized to the GI mucosa.10
axis.57 By contrast, T2Rs, a large family of receptor The understanding of the multiple pathways and
subtypes (more than 25 different genes in humans and systems involved in nutrient sensing can provide oppor-
more than 30 in rodents)58 detect a large group of bitter tunities to develop strategies for the treatment of food
substances and are believed to play a role in defending associated disorders by targeting specific EEC receptors.
the body from external threats.59 Distinct subtypes of
T2Rs have been localized in the GI mucosa, including
EECS AND THE BRAIN–GUT AXIS
subpopulations of EECs.52,54 Intraluminal bitter tas-
tants have been shown to induce release of ghrelin by
Brain–gut axis crosstalk
EECs45 and activate vagal neurons expressing CCK1
and Y2 receptors which bind to CCK and PYY, The relationship between the brain and the gut repre-
respectively, thus suggesting release of these peptides sents an important area of investigation in neurogas-
in response to bitter tastants.46 troenterology. The brain receives constant input of
different stimuli/signals from the GI tract and initiates
the appropriate integrated response to target cells in
Amino acids receptors
the GI tract.73 The information is predominantly
Different types of amino acids activate distinct receptors carried from the gut to the brain via vagal and spinal
on EECs in addition to the umami-T1R1-T1R3 sensing afferent neurons, immune mediators, gut hormones,
receptors2; examples include the Gq-coupled calcium and microbiota-related signaling molecules, while
sensing receptor (CaSR) that detects aromatic and basic information from the brain to the gut is via autonomic
aminoacids and is located in G, D, and I cells,60 and neurons and neuroendocrine factors.10,73 Although it is
GPRC6A and GPR92 that are activated by basic and now well established that there is bidirectional cross-
neutral aminoacids or protein hydrolysates, respectively, talk between the brain and the gut (Fig. 2) many
which are predominantly expressed by G and D cells.57,61 questions remain unsolved such as how the interplay
between the central nervous system (CNS) and the
periphery is perturbed in pathophysiological states. A
Free fatty acid receptors
typical example of brain–gut axis dysfunction is
G protein-coupled receptors which bind free fatty acid apparent in functional GI disorders, such as the
are involved in sensing medium and long-chain fatty irritable bowel syndrome and functional dyspepsia.74

© 2015 John Wiley & Sons Ltd 5


R. Latorre et al. Neurogastroenterology and Motility

Figure 2 EECs in the Brain–Gut Axis Bidirectional Communication. Green and Orange arrows indicate the bidirectional interplay referred to brain–
gut axis, which include the mucosal EECs, ENS, vagus nerve, spinal cord and CNS. The basal cytoplasmic processes have a role in activating intrinsic
and extrinsic sensory neurons. The activation of EECs may leads to sensitization via direct cell-to-neurons contact. Hormones released by EECs upon
stimulation act on receptors located on the vagal and spinal neurons and influence neuronal pathways in the bidirectional brain–gut communication.
Open type EECs sense luminal content and make contact with enteric nervous system (black), vagal nerve (afferent and efferent pathways, violet
lines) and spinal nerve (afferent and efferent pathways, blue lines), closed-type EEC also contribute to activate these feedback signals from the gut to
the brain. Together, this information sends positive or negative feedback to the brain (CNS), which modulates functions of the GI tract, glucose
homeostasis and satiety. Moreover, impairment in the brain–gut axis pathways could be implicated in obesity, visceral hypersensitivity, intestinal
disorders such as irritable bowel syndrome and inflammatory disorders such as inflammatory bowel disease. Abbreviations: NTS, nucleus tractus
solitarii; DRG, dorsal root ganglia; ENS, enteric nervous system (myenteric and submucosal plexus); EEC, enteroendocrine cell.

EECs secreting cholecystokinin Cholecystokinin is


EECs: role in brain–gut axis communication
mainly secreted in the upper small intestine in
The brain–gut axis is a multilevel system to manage response to fat and proteins.3,77 The postprandial
various aspects of GI physiology including nutrient release of CCK activates intestinal feedback through
signals and related central processing. Enteroendocrine extrinsic neural (vagal) pathway to control GI func-
cells contribute to these mechanisms by exerting regu- tions such as short-term inhibition of gastric emptying
latory effects on this bidirectional communication and acid secretion, through CCK1 receptors expressed
between the brain and the gut. Vagal afferent terminals by vagal afferent terminals that run in the lamina
innervate the wall of the GI tract, reaching the mucosa propria adjacent to the mucosal epithelium.75 Chole-
and the lamina propria and are in close proximity to the cystokinin can transmit sensory signals from the gut
mucosal epithelium.75 Peptides/hormones secreted by lumen by direct EEC-nerve communication and/or
EECs act on receptors located along the vagal afferent activation of vagal pathways through a paracrine
fibers. Vagal afferent pathways convey stimuli generated mechanism.21,75 Exogenous administration of CCK
by EECs to the brainstem, specifically the NTS, and the activates CCK1 receptors on vagal terminals and
nodose ganglion represents an intermediate station in induces satiation and reduction of meal size, while
the brain–gut axis bidirectional communication. There- the administration of CCK1 receptor antagonist
fore, vagal afferent pathways activated by peptides reverses these effects. Furthermore, exogenous admin-
released by EEC mediate numerous functions including istration of CCK or nutrients, such as protein or fat, in
the regulation of food intake, motility, secretion, CCK1 receptor null mice does not affect inhibition of
inflammatory response, and mucosal defense.73,76 food intake and gastric emptying, thus supporting the
Below are some examples of EEC-brain communication. role of CCK/CCK1 receptor in mediating these specific

6 © 2015 John Wiley & Sons Ltd


EEC cells in the brain-gut communication

effects.78 Moreover, in CCK1 receptor null mice there tion90 and reduced food intake.87 However, the short
was a strong decrease in activation of the vagal afferent half-life of GLP-1 due to the rapid breakdown results in
pathways in response to intestinal lipids compared to higher hormone concentration close to the site of
wild type mice, confirmed by lack of firing NTS secretion, and in pig intestine only a small part of that
neurons.78 The effect of CCK in the peripheral regula- reaches the systemic circulation before it is degraded by
tion of food intake is enhanced by leptin. When co- Dipeptidyl peptidase-IV.91 Thus, a question that arises,
administered, CCK and leptin increase the excitatory is whether GLP-1 physiological levels (bloodstream) in
response on vagal afferents, which leads to an exagger- human are capable of reaching and activating receptors
ated inhibition of gastric emptying and food intake.79 in the hypothalamus and brainstem. Glucagon-like
Moreover, the expression of leptin receptors on vagal peptide-1, secreted by EECs, acts as an endocrine
afferent neurons increase and decrease, respectively, in substance and also in a neural manner by activating
fasting or refeeding states.80 In addition to satiety, the receptors on the vagal afferent terminals close to the site
exogenous administration of CCK-8 in rats contributes of secretion, or receptors expressed by the ENS. Indeed,
to lower glucose production.81 The role of CCK1 the inhibition of food intake due to an intraperitoneal
receptor on vagal afferent terminals innervating the administration of GLP-1 in rodents is attenuated by the
small intestine is fundamental to activate the protein ablation of vagal–brainstem–hypothalamic pathways.87
kinase A signaling pathways involving the N-methyl- Prominent basal processes are frequently described in L-
D-aspartate receptor, which mediates neuronal trans- type EEC,20 which are likely involved in driving the
mission in the NTS to regulate glucose homeostasis.82 GLP-1 secretion directly on nerve terminals receptors as
‘real time’ response to the ingested nutrient present in
EEC secreting PYY Peptide YY is a 36 amino acid the gut lumen.
peptide produced mainly in the ileum and colon in Taken together, these data provide evidence sup-
response to feeding especially lipids, but also carbohy- porting the concept that hormones released from the
drates and proteins.32 Peptide YY is a member of the EECs activate vagal afferent pathways in a paracrine
neuropeptide Y (NPY) family of peptides which are manner or via direct EECs-sensory neuron communi-
expressed in different areas of the CNS, and five different cation, and therefore influence gut activity (e.g.,
NPY receptors (Y1, Y2, Y3, Y4, Y5) have been charac- intestinal motility, acid secretion, pancreatic enzyme
terized along the brain–gut axis.83 Peptide YY positive production and secretion, and gallbladder contraction)
neurons are found in the rodent NTS, and nerve termi- through the brain–gut axis.
nals containing PYY are localized in the pons, hypotha-
lamus (key energy balance regulating area in the brain),
EECs: role in sensory neural regulation
and spinal cord.84 The role of PYY in reducing food
intake is mainly due to the activation of Y2 receptor in A significant challenge in the study of EECs and their
the hypothalamus.85 However, exogenous administra- connection with epithelial cells and nerves is their
tion of PYY3-36 has been shown to exert effects on gastric sparse and irregular localization in the mucosal epithe-
emptying at least in part via the activation of vagal lium. Indirect methods, such as the measurement of
afferent pathways.86 The localization of Y2 receptor on plasma hormone levels in response to drugs or meals,
vagal afferent neurons support that PYY neurons and result in indirect evidence to support the physiological
nerve terminals in the vagus nerve convey visceral role(s) of EECs and are not helpful when addressing
sensory information from the gut to the brain and mechanisms of hormone release.76 Enteroendocrine
provide a central and peripheral control of food intake.87 cell lines are a useful tool to assess their role and
function, but an obvious limitation of isolated cell
EEC secreting GLP-1 Glucagon-like peptide-1, an lines is that they are not fully representative of the
incretin hormone, is released in the small intestine in more complex response seen in vivo.76 The develop-
response to food ingestion, especially carbohydrates and ment of transgenic mice tagged for hormone promoters
fats, and its main effect is on energy balance and blood driving the expression of GFP provides an excellent
glucose homeostasis.32 Glucagon-like peptide-1 recep- means to study the relationship of EECs with other
tor (GLP-1R) is peripherally distributed in the gut, cells, including sensory neurons. These studies have
pancreatic b-cells, kidney and in the vagus nerve and discovered that basal cytoplasmic processes in CCK-
centrally in the NTS and hypothalamus.88 Exogenous and PYY-GFP cells resemble axon with synaptic-like
stimulation of GLP-1R, peripherally or centrally, results ends containing most of the cell secretory vesicles,
in different physiological responses, such as inhibition many of which are distributed to the tips thus
of gastric emptying,89 glucose-stimulated insulin secre- suggesting they guide hormone secretion.8,20,92 These

© 2015 John Wiley & Sons Ltd 7


R. Latorre et al. Neurogastroenterology and Motility

basal cytoplasmic processes, which have been named SUMMARY AND CONCLUSION
‘neuropods’, contain intermediate filaments, are sur-
Enteroendocrine cells play an important role in gut
rounded by glia19 and appear to make a direct connec-
chemosensing to orchestrate appropriate functional
tion with nerves, including sensory nerve endings.20
responses to a variety of stimuli ranging from nutrients,
Furthermore, in vitro study showed a strong attraction
food degradation products, toxic chemicals, microorgan-
and subsequently connection between enteroendocrine
isms, and bacteria products. This complex balance
CCK-GFP cells and sensory neurons enzymatically
between maximizing nutrient assimilation and mini-
dissociated from the trigeminal nerves and from the
mizing danger to maintain optimal homeostasis is the
dorsal root ganglion of wild type mouse.20 Although
result of the interplay of different detection systems
these findings do not exclude other mechanisms of
including the distinct types of sensory receptors
action of EECs such as paracrine or through the
expressed by EECs, the variety of signaling molecules
bloodstream, they provide evidence for a direct con-
released by EECs and the targets that are activated
nection between EEC basal processes and mucosal
directly and indirectly such as neighboring cells, distant
nerves supporting the dual function of EECs as nutri-
targets reached via the bloodstream and neuronal path-
ent chemosensing at the top of the cell, while convey-
ways. Considerable progress has been made in under-
ing electrochemical information with cell-to-cell and/
standing EECs relationship with neurons of the ENS and
or cell-to-nerve connections at the bottom of the cell.
CNS and EEC bidirectional communication with the
brain. There is now strong evidence that EECs can
EECs: therapeutic potential directly participate in the brain–gut axis bidirectional
communication through axon-like processes or basal
The EEC-sensory neuron connection has thus opened a
cytoplasmic processes. The basal processes connect
new exciting prospective on EECs and their role in
EECs and neurons innervating the gut forming neuroep-
the communication with ENS and CNS. The direct
ithelial circuits, which allow sensory transmission from
cell-to-sensory nerves and neurons connections and the
the gut lumen and feedback from the brain to the gut
presence of presynaptic and postsynaptic genes in the
EECs. The activation of afferent and efferent nerve
basal cytoplasmic processes20 shed new light on our
pathways may be explored for the management of
understanding of the complexity of the bidirectional
diseases related to feeding disorders and GI dysfunction.
communication between the brain and the gut.
Alterations in the bidirectional communication
Enteroendocrine cell receptors on the luminal side
between the brain and the gut are likely associated with
could be a potential target of new drugs to activate
an impairment of gut functions (e.g., intestinal secre-
hormonal and neuronal pathways providing a novel
tion, motility, blood flow and afferent sensitivity). The
approach to treat diseases such as diabetes and obesity.
modification of the homeostatic reflex might lead to a
Currently, GLP-1R agonist is used to treat diabetes
chronic perturbation of the gut such as visceral hyper-
mellitus type 2, based on its function to stimulate
sensitivity. Therefore, a better understanding of the EEC
insulin secretion from pancreatic b-cells.93 Recently,
roles and their relationship with the brain and sensory
researchers have highlighted the beneficial role of GLP-
nerves is an important research target. Future studies are
1 and PYY3-36 secretion in the reduction of food intake
required to fully decipher this complex network. The
in patients undergoing gastric bypass surgery.94 Thus,
direct EEC to nerve connection opens a new horizon of
PYY and GLP-1 are now targets to treat obesity, a major
research and provides new tools to understand the
worldwide public health problem.95 Furthermore, drugs
mechanisms underlying EEC control of food intake and
acting to alter EEC functions may participate in the
GI dysfunction and potential new therapeutic targets.
control of depression, anxiety, and visceral hypersensi-
tivity, which are key components of functional GI
disorders.96 Recently, a significant amount of attention ACKNOWLEDGMENTS
has been given to a subgroup of EECs secreting 5-HT
The authors would like to thank the following funding agencies
and their role in visceral perception. 5-HT is a key for support: National Institutes of Health grants: P30 DK041301,
neurotransmitter in the control of nociceptive CURE Digestive Diseases Research Core Grant (CS), RO1-
responses and mood, with receptors located in both DK098447 (CS), UCLA Spitzer Award (CS), Department of
the periphery and CNS. Much evidence suggests that Veterans Affairs: Research Career Scientist Award to BG-VM
and VA Merit BX002188-01 (BG-VM) and VA Merit BX001195-05
activation of 5-HT receptors, specifically 5-HT3 and 5- (BG-VM). R. De G. is supported by grants from the Italian
HT4, sends signals from the gut to the CNS through Ministry of Public Health (Ricerca Finalizzata RER2009 - Ita-
vagal afferent fibers, which activate multiple pathways MNGIE); RFO from the University of Bologna; and ‘Fondazione
Del Monte di Bologna e Ravenna’.
including those involved in nociception.97–99

8 © 2015 John Wiley & Sons Ltd


EEC cells in the brain-gut communication

FUNDING DISCLOSURE
This review was not supported by any founding agency. The authors have no conflict of interest.

REFERENCES 14 Dockray GJ. Bayliss-Starling prize active products. Physiol Rev 1996;
lecture. Gastrointestinal hormones 76: 767–98.
1 Rehfeld JF. A centenary of gastroin- and the dialogue between gut and 25 Dockray GJ. The gut endocrine sys-
testinal endocrinology. Horm Metab brain. J Physiol 2014; 592: 2927–41. tem and its control. Exp Physiol 1994;
Res 2004; 36: 735–41. 15 Li HJ, Ray SK, Singh NK, Jhonston B, 79: 607–34.
2 Janssen S, Depoortere I. Nutrient Leiter AB. Basic helix-loop-helix tran- 26 Kending DM, Grider JR. Serotonin
sensing in the gut: new road to ther- scription factors and enteroendocrine and colonic motility. Neurogastroen-
apeutics? Trends Endocrinol Metab cell differentiation. Diabetes Obes terol Motil 2015; 27: 899–905.
2013; 24: 92–100. Metab 2011; 13: 5–12. 27 Gershon MD. 5-Hydroxytryptamine
3 Sternini C, Anselmi L, Rozengurt E. 16 Artavanis-Tsakonas S, Rand MD, (serotonin) in the gastrointestinal
Enteroendocrine cells: a site of Lake RJ. Notch signaling: cell fate tract. Curr Opin Endocrinol Diabetes
“taste” in the gastrointestinal control and signal integration in Obes 2013; 20: 14–21.
chemosensing. Cur Opin Endocri development. Science 1999; 284: 28 Keating DJ, Spencer NJ. Release of 5-
Diabetes Obes 2008; 15: 73–8. 770–6. Hydroxytryptamine from the mucosa
4 Larsson LI, Goltermann N, De Mag- 17 H€ofer D, Asan E, Drenckhahn D. is not required for the generation or
istris L, Rehfeld JF, Schwarz TW. Chemosensory perception in the gut. propagation of colonic migrating
Somatostatin cell processes as path- News Physiol Sci 1999; 14: 18–23. motor complexes. Gastroenterology
ways for paracrine secretion. Science 18 Gribble FM, Reimann F. Enteroen- 2010; 138: 659–70.
1979; 205: 1393–5. docrine cells: chemosensors in the 29 Spencer NJ, Nicholas SJ, Robinson L,
5 B€ottcher G, Sj€ olund K, Ekblad E, intestinal epithelium. Annu Rev Kyloh M, Flack N, Brookes SJ,
Hakanson R, Schwartz TW, Sundler Physiol 2016; 78: 3.1–3.23. Zagorodnyuk V, Keating DJ. Mecha-
F. Coexistence of peptide YY and 19 Bohorquez DV, Samsa LA, Roholt A, nisms of underlying distension-
glicentin immunoreactivity in endo- Medicetty S, Chandra R, Liddle RA. evoked peristalsis in guinea pig distal
crine cells of the gut. Regul Pept 1984; An enteroendocrine cell-enteric glia colon: is there a role for enterochro-
8: 261–6. connection revealed by 3D electron maffin cells? Am J Physiol Gastroin-
6 Keast JR, Furness JB, Costa M. Distri- microscopy. PLoS ONE 2014; 9: test Liver Physiol 2011; 301: G519–
bution of certain peptide-containing e89881. 27.
nerve fibers and endocrine cells in the 20 Bohorquez DV, Shaid RA, Erdmann 30 Mawe GM, Hoffman JM. Serotonin
gastrointestinal mucosa in five mam- A, Kreger AM, Wang Y, Calakos N, signaling in the gut-functions, dys-
malian species. J Comp Neurol 1985; Wang F, Liddle RA. Neuroephitelia functions and therapeutic targets.
236: 403–22. circuit formed by innervation of sen- Nat Rev Gastroenterol Hepatol
7 Boh orquez DV, Chandra R, Samsa LA, sory enteroendocrine cells. J Clin 2013; 10: 473–86.
Vigna S, Liddle RA. Characterization Invest 2015; 125: 782–6. 31 Klok MD, Jakobsdottir S, Drent ML.
of basal pseudopod-like processes in 21 Egerod KL, Engelstoft MS, Grunddal The role of leptin and ghrelin in the
ileal and colonic PYY cells. J Mol KV, Nøhr MK, Secher A, Sakata I, regulation of food intake and body
Histol 2011; 42: 3–13. Pedersen J, Windeløv JA et al. A weight in humans: a review. Obes
8 Chandra R, Samsa L, Vigna S, major lineage of enteroendocrine cells Rev 2007; 8: 21–34.
Liddle R. Pseudopod-like basal cell coexpress CCK, secretin, GIP, GLP-1, 32 Cummings DE, Overduin J. Gastroin-
processes in intestinal cholecys- PYY, and neurotensin but not testinal regulation of food intake. J
tokinin cells. Cell Tis Res 2010; 341: somatostatin. Endocrinology 2012; Clin Invest 2007; 117: 13–23.
289–97. 153: 5782–95. 33 Sakata I, Nakamura K, Yamazaki M,
9 Dockray GJ. Enteroendocrine cell sig- 22 Habib AM, Richards P, Cairns LS, Matsubara M, Hayashi Y, Kangawa K,
nalling via the vagus nerve. Curr Rogers GJ, Bannon CA, Parker HE, Sakai T. Ghrelin-producing cells
Opin Pharm 2013; 13: 954–8. Morley TC, Yeo GS et al. Overlap of exist as two types of cells, closed-
10 Furness JB, Riviera LR, Cho HJ, Bravo endocrine hormone expression in and opened type cells, in the rat
DM, Challagan B. The gut as a sen- the mouse intestine revealed by gastrointestinal tract. Peptides 2002;
sory organ. Nat Rev Gastroenterol transcriptional profiling and flow 23: 531–6.
Hepatol 2013; 10: 729–40. cytometry. Endocrynology 2012; 153: 34 Holst JJ. Incretin hormones and the
11 Psichas A, Reimann F, Gribble FM. 3054–65. satiation signal. Int J Obes (Lond)
Gut chemosensing mechanisms. J 23 Cho H-J, Kosari S, Hunne B, Cal- 2013; 37: 1161–8.
Clin Invest 2015; 125: 908–17. laghan B, Rivera LR, Bravo DM, Fur- 35 Sigalet DL, Wallace LE, Holst JJ,
12 Breer H, Eberle J, Frick C, Haid D, ness JB. Differences in hormone Martin GR, Kaji T, Tanaka H, Shar-
Widmayer P. Gastrointestinal localisation patterns of K and L type key KA. Enteric neural pathways
chemosensation: chemosensory cells enteroendocrine cells in the mouse mediate the anti-inflammatory
in the alimentary tract. Histochem and pig small intestine and colon. actions of glucagon-like peptide 2.
Cell Biol 2012; 138: 13–24. Cell Tissue Res 2015; 359: 693–8. Am J Physiol Gastrointest Liver
13 Mayer EA, Tillisch K, Gupta A. Gut/ 24 Dockray GJ, Varro A, Dimaline R. Physiol 2007; 293: G211–21.
brain axis and the microbiota. J Clin Gastric endocrine cells: gene expres- 36 Rowland KJ, Brubaker PL. The “cryp-
Invest 2015; 125: 926–38. sion, processing, and targeting of tic” mechanism of action of gluca-

© 2015 John Wiley & Sons Ltd 9


R. Latorre et al. Neurogastroenterology and Motility

gon-like peptide-2. Am J Physiol Gas- Integr Comp Physiol 2009; 296: ular receptive ranges of human
trointest Liver Physiol 2011; 301: G1– R528–36. TAS2R bitter taste receptors. Chem
8. 48 Glendinning JI, Yiin YM, Ackroff K, Senses 2010; 35: 157–70.
37 Hsieh J, Longuet C, Maida A, Bah- Sclafani A. Intragastric infusion of 60 MacLeod RJ. CaSR function in the
rami J, Xu E, Baker CL, Brubaker PL, denatonium conditions flavor aver- intestine: hormone secretion, elec-
Drucker DJ et al. Glucagon-like pep- sion and delays gastric emptying in trolyte absorption and secretion, para-
tide-2 increase intestinal lipid absorp- rodents. Physiol Behav 2008; 93: 757– crine non-canonical Wnt signaling
tion and chylomicron production via 65. and colonic crypt cell proliferation.
CD36. Gastroenterology 2009; 137: 49 Hagbom M, Istrate C, Engblom D, Best Pract Res Clin Endocrinol
997–1005. Karlsson T, Rodriguez-Diaz J, Buesa J, Metab 2013; 27: 385–402.
38 Batterham RL, Cohen MA, Ellis SM, Taylor JA, Loitto VM et al. Rotavirus 61 Haid DC, Jordan-Biegger C, Wid-
Le Roux CW, Withers DJ, Frost GS, Stimulates Release of Serotonin mayer P, Breer H. Receptors respon-
Ghatei MA, Bloom SR. Inhibition of (5-HT) from human enterochromaffin sive to protein breakdown products in
food intake in obese subjects by pep- cells and activates brain structures g-cells and d-cells of mouse, swine
tide YY3-36. N Engl J Med 2003; 349: involved in nausea and vomiting. and human. Front Physiol 2012; 3: 1–
941–8. PLoS Pathol 2011; 7: e1002115. 15. doi:10.3389/fphys.2012.00065.
39 Halatchev IG, Cone RD. Peripheral 50 Laffitte A, Neiers F, Briand L. Func- 62 Edfalk S, Steneberg P, Edlund H.
administration of PYY(3-36) produces tional roles of the sweet taste receptor Gpr40 is expressed in enteroen-
conditioned taste aversion in mice. in oral and extraoral tissues. Curr docrine cells and mediates free fatty
Cell Metab 2005; 1: 159–68. Opin Nutr Metab Care 2014; 17: 379– acid stimulation of incretin secretion.
40 Spiller RC, Trotman IF, Higgins BE, 85. Diabetes 2008; 57: 2280–7.
Ghetei MA, Grimble GK, Lee YC, 51 Behrens M, Meyerhof W. Oral and 63 Janssen S, Laermans J, Iwakura H,
Bloom SR, Misiewicz JJ et al. The extraoral bitter taste receptors. Tack J, Depoortere I. Sensing of fatty
ileal brake-inhibition of jejunal motil- Results Probl Cell Differ 2010; 52: acids for octanoylation of ghrelin
ity after ileal fat perfusion in man. 87–99. involves a gustatory G-protein. PLoS
Gut 1984; 25: 365–74. 52 Rozengurt N, Wu SV, Chen MC, ONE 2012; 7: 7e40168.
41 Giralt M, Vergara P. Glucagonlike Huang C, Sternini S, Rozengurt E. 64 Hirasawa A, Tsumaya K, Awaji T,
peptide-1 (GLP-1) participation in Colocalization of the alpha-subunit of Katsuma S, Adachi T, Yamada M,
ileal brake induced by intraluminal gustducin with PYY and GLP-1 in L Sugimoto Y, Miyazaki S et al. Free
peptones in rat. Dig Dis Sci 1999; 44: cells of human colon. Am J Physiol fatty acids regulate gut incretin gluca-
322–9. Gastrointest Liver Physiol 2006; 291: gon-like peptide-1 secretion through
42 Van Citters GW, Lin HC. Ileal brake: G792–802. GPR120. Nat Med 2005; 11: 90–4.
neuropeptidergic control of intestinal 53 Margolskee RF, Dyer J, Kokrashvili Z, 65 Brown AJ, Goldsworthy SM, Barnes
transit. Curr Gastroenterol Rep 2006; Salmon SH, Ilegems E, Daly K, Eme- AA, Eilert MM, Tcheang L, Daniels
8: 367–73. line L, Maillet EL et al. T1R3 and D, Muir AI, Wigglesworth MJ et al.
43 Daniel H, Zietek T. Taste and move: gustducin in gut sense sugar to regu- The orphan G protein-coupled recep-
glucose and peptide transporters in late expression of Na+-glucose tors GPR41 and GPR43 are activated
the gastrointestinal tract. Exp Physiol cotransporter. Proc Natl Acad Sci by propionate and other short chain
2015; 100: 1441–50. doi:10.1113/ USA 2007; 104: 15075–80. carboxylic acids. J Biol Chem 2003;
EP085029. 54 Vegezzi G, Anselmi L, Huynh J, 278: 11312–9.
44 Dockray GJ. Making sense of gut Barocelli E, Rozengurt E, Raybould 66 Karaki S, Tazoe H, Hayashi H, Kashi-
contents. Scand J Gastroenterol H, Sternini C. Diet-induced regula- wabara H, Tooyama K, Suzuki Y,
2003; 38: 451–5. tion of bitter taste receptor subtypes Kuwahara A. Expression of the
45 Janssen S, Laermans J, Verhulst PJ, in the mouse gastrointestinal tract. short-chain fatty acid receptor,
Thijs T, Tack J, Depoortere I. Bitter PLoS ONE 2014; 9: e107732. GPR43, in the human colon. J Mol
taste receptors and a-gustducin regu- 55 Li X, Staszewski L, Xu H, Durick K, Histol 2008; 39: 135–42.
late the secretion of ghrelin with Zoller M, Adler E. Human receptors 67 Tazoe H, Otomo Y, Karaki S, Kato I,
functional effects on food intake and for sweet and umami taste. Proc Natl Fukami Y, Terasaki M, Kuwahara A.
gastric emptying. Proc Natl Acad Sci Acad Sci USA 2002; 99: 4692–6. Expression of short-chain fatty acid
USA 2011; 108: 2094–9. 56 Nelson G, Hoon MA, Chandrashekar receptor GPR41 in the human colon.
46 Hao S, Sternini C, Raybould HE. Role J, Zhang Y, Ryba NJP, Zuker CS. Biomed Res 2009; 30: 149–56.
of CCK1 and Y2 receptors in activa- Mammalian sweet taste receptors. 68 Tolhurst G, Heffron H, Lam YS,
tion of hindbrain neurons induced by Cell 2001; 106: 381–90. Parker HE, Habib AM, Diakogian-
intragastric administration of bitter 57 Depoortere I. Taste receptors in the naki E, Cameron J, Grosse J et al.
taste receptor ligands. Am J Physiol gut tune the release of peptides in Short-chain fatty acids stimulate
Regul Integr Comp Physiol 2008; 294: response to nutrients. Peptides 2015; glucagon-like peptide-1 secretion
33–8. 66: 9–12. via the G- protein-coupled receptor
47 Hao S, Dulake M, Espero E, Sternini 58 Bachmanov AA, Beauchamp GK. FFAR2. Diabetes 2012; 61: 364–71.
C, Raybould HE, Rinaman L. Central Taste receptor genes. Annu Rev Nutr 69 Samuel BS, Shaito A, Motoike T, Rey
Fos expression and conditioned flavor 2007; 27: 389–414. FE, Backhed F, Manchester JK, Ham-
avoidance in rats following intragas- 59 Meyerhof W, Batram C, Kuhn C, mer RE, Williams SC et al. Effects of
tric administration of bitter taste Brockhoff A, Chudoba E, Bufe B, the gut microbiota on host adiposity
receptor ligand. Am J Physiol Regul Appendino G, Behrens M. The molec- are modulated by the short-chain

10 © 2015 John Wiley & Sons Ltd


EEC cells in the brain-gut communication

fatty-acid binding G protein-coupled F, Lewin MJ et al. Expression and GLP-1. Prog Mol Biol Transl Sci 2014;
receptor, Gpr41. Proc Natl Acad Sci regulation of leptin receptor protein 121: 23–65.
USA 2008; 105: 16767–72. in afferent and efferent neurons of the 91 Holst JJ, Deacon CF. Glucagone-like
70 Cani PD, Everard A, Duparc T. Gut vagus nerve. Eur J Neurosc 2001; 14: peptide-1 mediates the therapeutic
microbiota, enteroendocrine func- 64–72. actions of PP-IV inhibitors. Dia-
tions and metabolism. Curr Opin 81 Cheung GW, Kokorovic A, Lam CK, betologia 2005; 48: 612–5.
Pharmacol 2013; 13: 935–40. Chari M, Lam TK. Intestinal chole- 92 Boh orquez DV, Liddle RA. Axon-
71 Carabotti M, Scirocco A, Maselli MA, cystokinin controls glucose produc- like processes in enteroendocrine
Severi C. The gut-brain axis: interac- tion through a neuronal network. cells: characteristics and potential
tions between enteric microbiota, Cell Metab 2009; 10: 99–109. targets. Clin Transl Sci 2011; 4:
central and enteric nervous systems. 82 Rasmussen BA, Breen DM, Luo P, 387–91.
Ann Gastroenterol 2015; 28: 203–9. Cheung GWC, Yang CS, Sun B, 93 Meloni AR, De Young MB, Lowe C,
72 Nøhr MK, Pedersen MH, Gille A, Kokorovic A, Rong W et al. Duodenal Parkes DG. GLP-1 receptor activated
Egerod KL, Engelstoft MS, Husted AS, activation od cAMP-dependent pro- insulin secretion from pancreatic b-
Sichlau RM, Grunddal KV et al. tein kinase induces vagal afferent cell: mechanism and glucose depen-
GPR41/FFAR3 and GPR43/FFAR2 as firing and lower glucose production dence. Diabetes Obes Metab 2013;
cosensors for short-chain fatty acids in rats. Gastroenterology 2012; 142: 15: 15–27.
in enteroendocrine cells vs. FFAR3 in 834–43 e3. 94 Holst JJ. Enteroendocrine secretion of
enteric neurons and FFAR2 in enteric 83 Holzer P, Reichmann F, Farzi A. gut hormones in diabetes, obesity and
leukocytes. Endocrinology 2013; 154: Neuropeptide Y, peptide YY and pan- after bariatric surgery. Curr Opin
3552–64. creatic polypeptide in the gut-brain Pharmacol 2013; 13: 983–8.
73 Mayer EA. Gut feelings: the emerging axis. Neuropeptides 2012; 46: 261– 95 De Mello AH, Pra M, Cardoso LC, De
biology of gut–brain communication. 74. Bona Schraiber R, Rezin GT. Incretin-
Nat Rev Neurosci 2011; 12: 453–66. 84 Ekblad E, Sundler F. Distribution of based therapies for obesity treatment.
doi:10.1038/nrn3071. pancreatic polypeptide and peptide Metabolism 2015; pii: S0026-0495(15)
74 Mayer EA, Tillisch K. The brain- YY. Peptides 2002; 23: 251–61. 00155-9. doi: 10.1016/j.metabol.2015.
gut axis in abdominal pain syn- 85 Batterham RL, Cowley MA, Small CJ, 05.012.
dromes. Ann Rev Med 2011; 62: Herzog H, Cohen MA, Dakin CL, 96 Park JH, Rhee P-L, Kim G, Lee JH,
381–96. Wren AM, Brynes AE et al. Gut hor- Kim Y-H, Kim JJ, Rhee JC, Song SY.
75 Beyak M, Bulmer DCE, Jiang W, mone PYY(3-36) physiologically inhi- Enteroendocrine cell counts corre-
Keating C, Rong W, Grundy D. bits food intake. Nature 2002; 418: late with visceral hypersensitivity in
Extrinsic sensory afferent nerves 650–4. patients with diarrhea-predominant
innervating the gastrointestinal tract. 86 Whited KL, Tso P, Raybould HE. irritable bowel syndrome. Neurogas-
In: Johnson LR, Barrett KE, Ghishan Involvement of apolipoprotein A-IV troenterol Motil 2006; 18: 539–
FK, Merchant JL, Said HM, Wood JD, and CCk1 receptor in exogenous 46.
eds. Physiology of the Gastrointesti- PYY3-36-induced stimulation of 97 Greenwood-Van Meerveld B, Moham-
nal Tract, 4th edn. San Diego, CA: intestinal feedback. Endocrinology madi E, Tyler K, Pietra C, Bee LA,
Elsevier, 2006: 685–726. 2007; 148: 4695–703. Dickenson A. Synergistic effect of 5-
76 Raybould HE. Gut Chemosensing: 87 Abbott CR, Monteiro M, Small CJ, hydroxytryptamine 3 and neurokinin
interaction between gut endocrine Sajedi A, Smith KL, Parkinson JR, 1 receptor antagonism in rodent mod-
cells and visceral afferents. Auton Ghatei MA, Bloom SR. The inhibitory els of somatic and visceral pain. J
Neurosci 2010; 153: 41–6. effects of peripheral administration of Pharmacol Exp Ther 2014; 351: 146–
77 Dockray GJ. Luminal sensing in the peptide (3-36) and glucagon-like pep- 52.
gut: an overview. J Physiol Pharmacol tide-1 on food intake are attenuated by 98 Gilet M, Eutamene H, Han H, Kim
2003; 54: 9–17. ablation of the vagal-brainstem- HW, Bueno L. Influence of new 5-HT4
78 Whited KL, Thao D, Lloyd KC, Kopin hypothalamic pathway. Brain Res receptor partial agonist, YKP10811,
AS, Raybould HE. Targeted distrup- 2005; 1044: 127–31. on visceral hypersensitivity in rats
tion of the murine CCK1 receptor 88 Holst JJ. The physiology of glucagon- triggered by stress and inflammation.
gene reduces intestinal lipid-induced like peptide 1. Physiol Rev 2007; 87: Neurogastrenterol Motil 2014; 26:
feedback inhibition of gastric func- 1409–39. 1761–70.
tion. Am J Physiol Gastrointest Liver 89 _
Imery€ uz N, Yegen BC, Bozkurt A, 99 Keszthelyi D, Troost FJ, Jonkers DM,
Physiol 2006; 291: G156–62. Cosßkun T, Villanueva-Pe~ nacarrillo Van Eijk HM, Dekker J, Buurman
79 Peters JH, Simasko SM, Ritter RC. ML, Ulusoy NB. Glucagon-like pep- WA, Masclee AAM. Visceral hyper-
Modulation of vagal afferent excita- tide-1 inhibits gastric emptying via sensitivity in irritable bowel syn-
tion and reduction of food intake by vagal afferent-mediated central mech- drome: evidence for involvement of
leptin and cholecystokinin. Physiol anisms. Am J Physiol 1997; 273: serotonin metabolism- preliminary
Behav 2006; 89: 477–85. G920–7. study. Neurogastroenterol Motil
80 Buyse M, OvesjO € ML, Go€ıo H, Guil- 90 Nadkarni P, Chepurny OG, Holz GG. 2015; 27: 1127–37.
meau S, P eranzi G, Moizo L, Walker Regulation of glucose homeostasis by

© 2015 John Wiley & Sons Ltd 11

View publication stats

Vous aimerez peut-être aussi