ArticlePDF Available

Evidence for Neuronal and Structural Changes in Submucous Ganglia of Patients With Functional Dyspepsia

Authors:

Abstract and Figures

An intact and well-functioning enteric nervous system is necessary to efficiently organize gut function. Functional gastrointestinal disorders are pathological entities in which gut function is impaired without a clearly established pathophysiology. On the basis of the relative ease with which intestinal biopsies can be obtained, and taking advantage of a recently developed optical recording technique, we evaluated whether functional neuronal defects exist in enteric nerves of patients with functional dyspepsia (FD). The submucous plexus isolated from duodenal biopsies taken from FD patients and control subjects was used to functionally and morphologically examine nerves and ganglionic architecture (neurons and glial cells). In light of previous studies reporting eosinophil and mast cell infiltration in the gut mucosa of FD patients, we also examined whether these cells infiltrated the submucous plexus and whether this correlated with neuronal activity and specific clinical symptoms. We demonstrate that neuronal functioning is impaired in the submucous plexus of FD patients, as shown by decreased calcium responses to depolarization and electrical stimulation. Glial (S100) and neuronal (HuCD) markers show signs of gliosis, altered ganglionic architecture, and neuronal abnormalities in the submucous plexus of FD patients. We found that eosinophils and mast cells infiltrated the submucous layer of FD patients to a much larger extent than in controls. A significant correlation was found between the number of these cells and the calcium transient amplitudes measured in submucous ganglia. We provide the first direct evidence that FD is characterized by functional and structural abnormalities within the submucous ganglion plexus, which may be of future predictive and diagnostic value in the treatment of FD patients.Am J Gastroenterol advance online publication, 16 June 2015; doi:10.1038/ajg.2015.158.
Content may be subject to copyright.
see related editorial on page x
© 2015 by the American College of Gastroenterology The American Journal of GASTROENTEROLOGY
nature publishing group 1205
FUNCTIONAL GI DISORDERS
ORIGINAL CONTRIBUTIONS
INTRODUCTION
In up to 50% of patients seen in gastroenterology outpa-
tient clinics, routine investigations fail to identify underlying
abnormalities that can explain their symptoms and com-
plaints. Because of that reason, these patients are referred to as
having functional gastrointestinal disorders (FGIDs) ( 1,2 ),
comprising a set of diseases that, due to their chronic nature
and high prevalence, constitute a major personal and societal
burden ( 1–3 ).
Recent evidence points the attention to subtle cellular and
molecular alterations in the gastrointestinal tract of patients with
FGIDs ( 4–10 ). Although the presence of so-called ‘low-grade
in ammation’ together with abnormalities at the mucosal level
appear to be key features in patients with FGIDs, they do not fully
justify or explain the presence of (speci c) clinical symptoms
in these patients ( 1 ). Because of its crucial role in the control of
gut sensorimotor functions, the enteric nervous system has been
hypothesized to also have its part in the pathophysiology of FGIDs
Evidence for Neuronal and Structural Changes in
Submucous Ganglia of Patients With Functional
Dyspepsia
Carla Cirillo , MSc, PhD
1
,
2
, Talat Bessissow , MD, PhD
2
,
3
, An-Sofi e Desmet , MSc
1
,
2
, Hanne Vanheel , MSc, PhD
2
,
Jan Tack , MD, PhD
2
and Pieter Vanden Berghe , MSc, PhD
1
,
2
OBJECTIVES:
An intact and well-functioning enteric nervous system is necessary to effi ciently organize gut
function. Functional gastrointestinal disorders are pathological entities in which gut function is
impaired without a clearly established pathophysiology. On the basis of the relative ease with which
intestinal biopsies can be obtained, and taking advantage of a recently developed optical recording
technique, we evaluated whether functional neuronal defects exist in enteric nerves of patients with
functional dyspepsia (FD).
METHODS: The submucous plexus isolated from duodenal biopsies taken from FD patients and control subjects
was used to functionally and morphologically examine nerves and ganglionic architecture (neurons
and glial cells). In light of previous studies reporting eosinophil and mast cell infi ltration in the gut
mucosa of FD patients, we also examined whether these cells infi ltrated the submucous plexus and
whether this correlated with neuronal activity and specifi c clinical symptoms.
RESULTS: We demonstrate that neuronal functioning is impaired in the submucous plexus of FD patients, as
shown by decreased calcium responses to depolarization and electrical stimulation. Glial (S100)
and neuronal (HuCD) markers show signs of gliosis, altered ganglionic architecture, and neuronal
abnormalities in the submucous plexus of FD patients. We found that eosinophils and mast cells
infi ltrated the submucous layer of FD patients to a much larger extent than in controls. A signifi cant
correlation was found between the number of these cells and the calcium transient amplitudes
measured in submucous ganglia.
CONCLUSIONS: We provide the fi rst direct evidence that FD is characterized by functional and structural
abnormalities within the submucous ganglion plexus, which may be of future predictive and
diagnostic value in the treatment of FD patients.
Am J Gastroenterol 2015; 110:1205–1215; doi: 10.1038/ajg.2015.158; published online 16 June 2015
1
Laboratory for Enteric NeuroScience (LENS), University of Leuven , Leuven , Belgium ;
2
Translational Research Center for GastroIntestinal Disorders (TARGID),
University of Leuven , Leuven , Belgium ;
3
Department of Gastroenterology, Royal Victoria Hospital, McGill University Health Center , Montreal , Quebec , Canada .
Correspondence: Pieter Vanden Berghe, MSc, PhD, Laboratory for Enteric Neuroscience (LENS), TARGID, University of Leuven , Herestraat 49, O&N1 mailbox
701 , Leuven 3000 , Belgium . E-mail: pieter.vandenberghe@med.kuleuven.be
Received 29 December 2014 ; accepted 20 April 2015
The American Journal of GASTROENTEROLOGY www.amjgastro.com
1206
FUNCTIONAL GI DISORDERS
VOLUME 110
|
AUGUST 2015
Cirillo et al.
( 1,11–13 ). Although highly valuable, the current evidence for
this involvement is at best indirect as it is based on clinical and
basic pharmacological studies ( 14–19 ).  e few studies that used
intestinal biopsies to address the possibility of cellular defects
have focused their attention mostly on the mucosal layer ( 8–10 ),
whereas only two ( 7,20 ) also included the nerve plexus and quanti-
ed neuronal numbers.  e latter point is of extreme importance,
as neuronal quanti cation is most likely not su cient to identify
enteric neuropathies’ ( 1 ). We and others have recently demon-
strated that intestinal biopsies obtained during routine endoscopy
are a source of invaluable biological material, as they contain snip-
pets of submucous plexus, with ganglia and neurons that can be
quanti ed and recorded from ( 21–23 ).
Taking advantage of the recently developed optical recording
techniques ( 21 ) and the ease to obtain biopsies without major risks
( 24 ), we are now able to directly test the hypothesis that nerves in
the intestinal wall of patients with FGIDs are dysfunctional even
when gross morphology seems una ected ( 1 ). We thus used this
advanced optical approach in patients with functional dyspepsia
(FD), which is one of the most prevalent FGIDs. FD is de ned by
Rome III consensus as the presence of symptoms in the absence of
any underlying organic disease and whose pathophysiology is not
clearly established ( 1,25,26 ). Moreover, on the basis of the recent
evidence that low-grade in ammation may have a crucial role in
FD pathophysiology ( 8–10 ) and the fact that immune cells inter-
act with enteric nerves to in uence intestinal function ( 27 ), we
also evaluated how immune cell in ltration in the submucous
plexus of FD patients correlated with both neuronal activity and
the presence of speci c FD symptoms.
MATERIALS AND METHODS
Study population
In this study, we used duodenal biopsies from 18 newly diag-
nosed FD patients ful lling the Rome III criteria ( 2 ) who were
referred to our gastroenterology unit for endoscopic evaluation.
ese patients were invited to complete the Rome III dyspep-
sia module questionnaire ( 2,25,26 ). To facilitate diagnostic and
therapeutic approach to FD patients, the Rome III consensus pro-
posed to subdivide FD into the postprandial distress syndrome
(PDS), characterized by meal-related symptoms, such as early
satiation and postprandial fullness, and the epigastric pain syn-
drome (EPS), characterized by epigastric burning and epigastric
pain ( 2 ). EPS and PDS symptoms were thus assessed. In line with
Rome III criteria, we excluded patients with diabetes or celiac dis-
ease. As controls, we used 20 age- and sex-matched subjects who
were referred to our gastroenterology unit for endoscopic evalu-
ation for other reasons.  ese subjects will be termed ‘controls
throughout the rest of the manuscript.  ey had no macroscopic
upper gastrointestinal lesions at endoscopy and did not ful ll the
Rome III criteria for FD. All subjects underwent careful history
taking, clinical examination, and routine biochemistry together
with histological examination of duodenal biopsies. Exclusion
criteria for all participants were intake of non-steroidal anti-
in ammatory drugs, proton-pump inhibitors, corticosteroids, or
other immunosuppressive drugs in the 6 months prior to exami-
nation. All FD patients and controls were asked about food aller-
gies or intolerance.  e study protocol (ML7400) was approved
by the Ethics Committee of Leuven University Hospitals. Written
informed consent was obtained from each subject prior to inclu-
sion in the study.
Preparation, loading, and calcium (Ca
2+
)-imaging of submucous
neurons
Duodenal biopsies were taken by experienced endoscopists
(TB, JT) using standard biopsy forceps and put in ice-cold Krebs
solution (in mM: 120.9 NaCl, 5.9 KCl, 1.2 MgCl
2
, 2.5 CaCl
2
, 11.5
glucose, 14.4 NaHCO
3
, and 1.2 NaH
2
PO
4
) ( 21 ). A er removal,
biopsies were quickly transferred to the lab, and the mucosa
was removed by using watchmaker’s forceps to obtain the sub-
mucous plexus. All dissection procedures were performed in
ice-cold Krebs solution to reduce cellular activity and release of
mediators. A er loading (at room temperature) with the  uores-
cent Ca
2+
indicator Fluo-4 AM (Molecular Probes, Invitrogen,
Merelbeke, Belgium) and 0.01% Cremophor EL surfactant agent
(Fluka Chemika, Buchs, Switzerland), Ca
2+
-imaging was per-
formed as previously described ( 21 ). Fiber tracts were electrically
stimulated via a platinum electrode (diameter 50 μ m), which
was guided by a mechanical micromanipulator (Narishige,
London, UK). Trains (2 s, 20 Hz) of 300 μ s electrical pulses
(Grass Instruments, Quincy, MA) were used. Neurons were only
included in the Ca
2+
analysis when they displayed a sharply increas-
ing Ca
2+
response to high-K
+
(75 mM) perfusion. Images were
collected using TILL Vision so ware (TILL Photonics, Gräfel ng,
Germany), and analysis was performed using custom-written mac-
ros in IGOR PRO (Wavemetrics, Lake Oswego, OR). To remove
dri and movement artefacts due to perfusion, the image stack was
registered to the  rst image. Regions of interest were drawn over
each cell,  uorescence intensity was normalized to the basal
uorescence at the onset of the recording for each region of inter-
est (Δ F/F
0
), and peaks were analyzed. A peak was considered
if the signal rose above baseline plus  ve times the intrinsic
noise level.  e percentage of responsive cells and the maxi-
mum intracellular Ca
2+
concentrations peak amplitude were
determined. All experiments and analyses were performed in a
blinded way.
Immunofl uorescent staining of the submucous plexus
A er xation with paraformaldehyde, submucous plexus prepa-
rations were stained using primary antibodies against neuronal
and glial components (chicken anti-neuro lament 200 kDa
(NF200, 1:500; Abcam, Cambridge, UK), mouse anti-panneu-
ronal HuCD (1:500; Molecular Probes, Invitrogen), rabbit anti-
vasoactive intestinal peptide (VIP, 1:500, Millipore, Overijse,
Belgium), rabbit anti-S100 (1:500; Dako, Glostrup, Denmark)),
eosinophils and mast cells using mouse anti-eosinophilic major
basic protein (1:20; AbD Serotec, Kidlington, UK), and mouse
anti-mast cell tryptase (1:200; Dako). Primary antibody incuba-
tion was followed by a labeling step with appropriate  uorescently
labeled secondary antibodies. Immunohistochemical staining was
© 2015 by the American College of Gastroenterology The American Journal of GASTROENTEROLOGY
1207
FUNCTIONAL GI DISORDERS
Submucous Defects in Functional Dyspepsia
visualized under an epi uorescence microscope (BX 41 Olym-
pus, Belgium) with speci c lter cubes (EX/DM/EM in nm) for
blue (325–375/400/435–485), green (460–495/505/510–550),
red (570–590/595/600–660), and far red (620–650/655/660–720)
uorescent probes. Images were recorded using Cell^F so ware
on an XM10 (Olympus) camera. Each preparation was entirely
screened to count ganglia, neurons per ganglion, and also non-
ganglionic (individual) neurons, as described earlier ( 21 ). Confo-
cal images were recorded using Zeiss LSM 510 Meta and LSM 780
confocal microscopes (Cell Imaging Core, KU Leuven, Belgium).
Eosinophils and mast cells were counted in the whole submucous
plexus, and their number was expressed as a number of positive
cells/total surface of submucous plexus.
Analysis of HuCD, S100, and VIP staining in submucous
neurons
As described in an earlier paper ( 28 ), the localization of the HuCD
protein, used to identify neurons, can either be cytosolic or be
predominantly nuclear. In normal conditions, the HuCD protein
is homogeneously spread in the neuronal cell body, whereas in
conditions of experimental neuronal damage HuCD appears to be
present mainly within the nucleus ( 28 ). Each of the samples was
entirely screened, and dual color images were taken of each gan-
glion to evaluate the HuCD staining in a blinded way. Cells that
had a strong nuclear HuCD staining were counted, and the per-
centage over total neurons per ganglion was made for each biopsy.
Similarly, also the mean HuCD intensity signal was measured for
nucleus and cytosol of each neuron (Cell^F so ware, Olympus),
and the mean intensity nucleus/cytosol ratio was calculated for
each biopsy. For volumetric measurements of S100 expressing
cells, we used confocal recordings, which were deconvolved with
Huygens Professional (Scienti c Volume Imaging, Hilversum,
e Netherlands) and visualized in Imaris 7.7 (Bitplane, Zürich,
Switzerland) on an image analysis workstation (Cell Imaging
Core, KU Leuven, Belgium). Cubes of interest were drawn around
individual ganglia, and S100 positive voxels were determined in a
blinded way using the spot detection algorithm available in Imaris
7.7.  e total glial volume (in μ m
3
) was calculated and expressed
relative to the number of HuCD-positive neurons present in the
same cube of interest. For VIP analysis, we used a far red (Alexa
647) secondary antibody to avoid spectral overlap with the other
antibody staining. In order to assess VIP staining, all biopsies
were scored in a blinded manner, and  ber intensity was quanti-
ed in parallel using IGOR PRO (Wavemetrics).
Data and statistical analysis
All results are presented as mean±s.e.m.  e normal distribution
of the data was veri ed by the Kolmogorov–Smirnov test for nor-
mality. Di erences between groups were analyzed using two-tailed
unpaired Student’s t -test or the Mann–Whitney U -test. e num-
ber of neurons/ganglion and ganglia/biopsy was compared using
Fisher’s exact test.  e success rate for Ca
2+
-imaging in FD patients
and in controls was evaluated by the
χ
2
test.  e di erence in the
number (in %) of neurons responding to high-K
+
or electrical
stimulation (ES) was evaluated using Fisher’s exact test.  e cor-
relations between neuronal amplitudes and immune cell numbers
and EPS or PDS symptoms with neuronal amplitudes or immune
cell numbers were performed using Spearmans rank analysis.
P -values <0.05 were considered signi cant. Statistical analysis
was performed with Microso Excel 2007 (Redmond, WA),
GraphPad Prism 5.0 (GraphPad, San Diego, CA), or SAS enterprise
(Cary, NC).
RESULTS
Study population
Duodenal biopsies taken from 18 FD patients ful lling the Rome
III criteria were included in the study, 10 with EPS and 8 with
PDS ( 2,25,26 ), with 6 of them having overlapping characteristics.
Demographic and clinical characteristics, as well as the indica-
tions for gastro-duodenal endoscopy in FD patients and controls
are summarized in Table 1 . No signi cant di erences in age and
sex distribution were found between controls and FD patients.
None of the participants were taking medication (proton-pump
inhibitors, non-steroidal anti-in ammatory drugs, corticoster-
oids) in the 6 months prior to the examination, none were smok-
ers, and none had food allergies or intolerance.  e endoscopic
examination revealed no macroscopic changes in the duodenum
of any of the participants. All subjects were Helicobacter pylori
negative, and clinical examination and routine biochemistry
were not indicative of any organic disease. Routine histology of
Table 1 . Demographic and clinical characteristics of the study
subjects
Controls ( n =20) FD patients
( n =18)
P value
Gender (F:M) 12:8 12:6
Age, (range) 43.9±2.7, (22–74) 40.4±3.3,
(26–64)
0.33
a
Indication for
endoscopy ( n )
Anemia (7), follow-up
gastric ulcer (3), weight
loss (6), pre-operative
evaluation for gastro-
esophageal refl ux
disease (4)
FD
b
(18)
Rome III classifi -
cation
b
(EPS:PDS)
10:8
(overlap in 6)
Macroscopic
endoscopy
(duodenum)
Normal Normal
Routine histology
c
(duodenum)
Normal Normal
EPS, epigastric pain syndrome; F, female; FD, functional dyspepsia; M, male;
PDS, postprandial distress syndrome.
a
Mann–Whitney U -test.
b
Rome III criteria: postprandial fullness, early satiety, epigastric pain, or epigas-
tric burning ( 2,25,26 ).
c
Routine histology performed by the pathology department at Leuven University
Hospitals.
The American Journal of GASTROENTEROLOGY www.amjgastro.com
1208
FUNCTIONAL GI DISORDERS
VOLUME 110
|
AUGUST 2015
Cirillo et al.
duo denal biopsies did not reveal any microscopic change sug-
gestive of duodenopathy.  ere were no signi cant di erences
between FD patients and controls in terms of how easily the nerve
layer (submucous plexus) could be dissected and loaded with Ca
2+
indicator.
Quantitative analysis of enteric neurons and ganglia
Immunohistochemical analysis ( Figure 1a ) did not reveal any
di erence in the number of ganglia per biopsy in FD patients
compared with controls (7.9±1 vs. 7.3±1, respectively; P =0.63,
Figure 1b ). In addition, no changes were found in the number
of neurons per ganglion (5.0±0.1 vs. 4.8±0.1, P =0.09, Figure 1c )
nor in the number of non-ganglionic neurons (2.8±0.6 vs.
2.5±0.6, P =0.70, Figure 1d ) between FD patients and controls,
respectively.
Depolarization and electrically induced responses differ
between FD patients and controls
C a
2+
-imaging recordings were performed in 236 neurons from
49 ganglia of controls and 230 neurons from 48 ganglia of FD
patients, as recognized based on morphology and characteris-
tic Fluo-4 loading ( 21 ) ( Figure 2a and c ). Viable neurons were
identi ed with a brief high-K
+
depolarization. In controls, 76%
of the neurons responded to high-K
+
( Figure 2e , right), with an
average Ca
2+
transient amplitude of 8±1% ( Figure 2b and e ). In
FD patients, however, only 56% of neurons responded ( P <0.01,
Figure 2e , right), and their peak amplitude was signi cantly
lower (3±2%, P <0.01, Figure 2d and e ) compared with control
neurons.
Trains of electrical pulses applied to the interconnecting  ber
bundles (ES) activated 56% of the high-K
+
responsive neurons
in controls ( Figure 2e , right), with Ca
2+
transient amplitudes
reaching 5±0.2% ( Figure 2b and e ). In FD patients, only 28% of
the identi ed neurons responded to electrical stimuli ( P <0.01,
Figure 2e , right), and lower Ca
2+
transient amplitudes were
measured in responding neurons (2±0.1%, P <0.01, Figure 2d
and e ) compared with controls.
To investigate whether Ca
2+
response characteristics could
be associated with subpopulations of patients, we subdivided
the FD patients (in EPS and PDS) but did not  nd any signi -
cant di erence either a er high-K
+
depolarization (3±1 vs. 3±1%,
P =0.42) or a er electrical stimulation (1±0.2 vs. 1±0.2%, P =1.0;
Figure 2f ).
Presence of eosinophils and mast cells in the submucous
plexus of FD patients
Immunostaining of the submucous plexus for eosinophilic major
basic protein revealed a signi cantly higher number of eosino-
phils in FD patients compared with controls (112.8±13.5 vs.
10.2±2.8, P <0.01, total counts per layer, Figure 3a and b ), with
Submucous plexus
NF200
HuCD
NS NS NS
20
bcd
a
15
10
Ganglia/biopsy
Neurons/ganglion
5
0
10
8
6
4
0
2
Isolated neurons/biopsy
10
8
6
4
0
2
Controls Controls FD patients Controls FD patientsFD patients
S100
Merge
Figure 1 . The number of neurons and ganglia does not differ in functional dyspepsia (FD) patients compared with controls. ( a , left) Schematic representa-
tion of the submucous plexus (~5 mm
2
) isolated from duodenal biopsies and ganglia containing neurons (color code: green and red) and glial cells (color
code: blue) with interconnecting fi bers (color code: red and blue). ( a , right) Immunostaining of a submucous ganglion where neurons (HuCD, green), glial
cells (S100, blue), and nerve fi bers (NF200, red) can be identifi ed. Bar = 20 μ m. ( b ) Number of submucous ganglia per biopsy in FD patients compared
with controls. No signifi cant differences were found. ( c ) Number of neurons per ganglion in FD patients compared with controls. No signifi cant differences
were found. ( d ) Number of isolated neurons in the submucous plexus of FD patients compared with controls. No signifi cant differences (NS) were found.
© 2015 by the American College of Gastroenterology The American Journal of GASTROENTEROLOGY
1209
FUNCTIONAL GI DISORDERS
Submucous Defects in Functional Dyspepsia
of eosinophils and mast cells could be found in close vicinity
(<100 μ m) to neurons ( Figure 3a and c ).
When we compared the total number of in ltrating cells in the
submucous plexus, we found no signi cant di erences in the total
number of eosinophils in PDS compared with EPS (132.9±23.8 vs.
99.6±16.6, respectively, P =0.17; Figure 3e , le ), nor in the total
no di erences between symptom-based subgroups. Similarly,
when stained for the mast cell marker tryptase, the number of
positive cells was signi cantly higher in the submucous plexus
of FD patients compared with controls (223.6±28.2 vs. 69.2±6.0,
P <0.01, total counts per layer, Figure 3c and d ). Double-label
immunostaining using HuCD revealed that a signi cant number
20
15
20
30
10
5
0
15
20
10
5
0
3
4
5
2
1
0
EPS PDS EPS PDS
NS NS
**
**
**
**
8
4
6
10
2
0
4
2
3
5
1
0
4
2
3
5
1
0
Controls FD patients
Controls FD patients
Controls
High K+
High K
+
High K
+
ES
ES
ES
FD patients
Controls
FD patients
15
20
ab
c
e
f
d
High K
+
ES
20 40
Seconds Seconds
60 80 100
10
ΔF/F
0
(%)ΔF/F
0
(%)
ΔF/F
0
(%)
ΔF/F
0
(%)
ΔF/F
0
(%)
ΔF/F
0
(%)
ΔF/F
0
(%)ΔF/F
0
(%)
Controls
1
1
2
2
5
0
0
20 40
Seconds
60 80 100
0
10 20 30 40 60500
10 20 30 40 6050
0
3
4
5
2
1
0
80
40
60
100
20
0
Percentage of
responding neurons
FD patients
2
1
1
2
Figure 2 . Neuronal impairment in the submucous plexus of functional dyspepsia (FD) patients. ( a ) Immunostaining of a typical ganglion (left) from control
and its matched Fluo-4 signal (right). ( b , left) Recordings of the responses of neuron 1 (red arrowhead) and 2 (green arrowhead) to high-K
+
showing
the typical fast upstroke. ( b , right) Recordings of the responses of neuron 1 (red arrowhead) and 2 (green arrowhead) to electrical stimulation (ES).
( c ) Immunostaining of a ganglion (left) from a FD patient and its matched Fluo-4 signal (right). Note that the neurons labeled express a brighter HuCD
staining in the nucleus (green). ( d , left) Recordings of the responses of neuron 1 (red arrowhead) and 2 (green arrowhead) to high-K
+
showing lower
amplitude compared with control neurons. ( d , right) Recordings of the responses of neuron 1 (red arrowhead) and 2 (green arrowhead) to ES: also in this
case, the amplitude is lower compared with control neurons. The arrowheads indicate the neurons of which Ca
2+
responses were plotted in the graphs.
The gray bars indicate the period of exposure to high-K
+
. ( e ) Relative fl uorescence (Δ F/F
0
) recorded from neurons when stimulated with high-K
+
(left)
and ES (middle). Each dot is the average response from neurons in the biopsies from controls (blue dots) and FD patients (red dots). ( e , right) Number
of neurons responding to high-K
+
and ES in FD patients (red bars) compared with controls (blue bars). Each bar shows the percentage of responding
neurons (identifi ed by high-K
+
depolarization) (** P < 0.01). ( f ) Relative fl uorescence (Δ F/F
0
) recorded from neurons when stimulated with high-K
+
(left) and
ES (right) in epigastric pain syndrome (EPS) and postprandial distress syndrome (PDS) subgroups. Each dot is the average response from neurons in the
biopsy. Bars = 20 μ m. NS, not signifi cant.
The American Journal of GASTROENTEROLOGY www.amjgastro.com
1210
FUNCTIONAL GI DISORDERS
VOLUME 110
|
AUGUST 2015
Cirillo et al.
number of mast cells per submucous plexus (EPS: 226.2±29.8 vs.
PDS: 242±55.7, P =0.87) ( Figure 3e , right).
Correlation between neuronal impairment and eosinophil/mast
cell number in FD patients and controls
To link the two main  ndings of this study, we evaluated whether
impairment of neuronal activation in the submucous plexus
correlated with the increased number of in ltrating eosinophils
and mast cells. To do so, we performed a Spearman correlation
test using the data generated by the Ca
2+
-imaging experiments
and immunohistochemical assessment independently of the clin-
ical classi cation ( Figure 4 ). We found a signi cant non-linear
negative correlation between the response amplitudes and the
amount of in ltrated cells, both for responses elicited by high-
K
+
depolarization (eosinophils: rho=−0.48, P =0.003; mast cells:
rho =−0.34, P =0.042, Figure 4a and b , respectively) or electrical
pulses (eosinophils: rho=−0.56, P =0.001; mast cells: rho =−0.52,
P =0.002, Figure 4c and d , respectively).  e Spearman test did
Controls
**
200
300
a
c
e
FD patients
Controls
FD patients
0
100
EPS PDS
200
Eosinophil cell number
300
0
200
EPS PDS
400
Mast cell number
600
Controls
MBP
MBP
HuCD
HuCD
100
0
Eosinophil cell number
FD patients
Controls FD patients
**
NS NS
Tryptase
HuCD
Tryptase HuCD
200
300
100
0
Mast cell number
Figure 3 . The number of eosinophils and mast cells in the submucous plexus is higher in functional dyspepsia (FD) patients than in controls. ( a ) Three
consecutive confocal images (2 μ m apart) of the submucous plexus of control (upper panels) and FD patients (lower panels) where neurons (HuCD, green)
and eosinophils (major basic protein (MBP), color code: magenta) can be identifi ed by immunohistochemistry. ( b ) Number of eosinophils in the
sub mucous plexus of FD patients compared with controls. ( c ) Three consecutive confocal images (2 μ m apart) of the submucous plexus of control
(upper panels) and FD patients (lower panels) where neurons (HuCD, green) and mast cells (tryptase, color code: red) can be identifi ed by immuno-
histochemistry. White arrows indicate immune cells; arrowheads indicate neurons. Bar = 20 μ m. ( d ) Number of mast cells in the submucous plexus of
FD patients compared with controls. ( e ) Number of eosinophils (left) and mast cells (right) in the submucous plexus of epigastric pain syndrome (EPS)
compared with postprandial distress syndrome (PDS) subgroups. ** P < 0.01; NS, not signifi cant.
© 2015 by the American College of Gastroenterology The American Journal of GASTROENTEROLOGY
1211
FUNCTIONAL GI DISORDERS
Submucous Defects in Functional Dyspepsia
of eosinophils was correlated to the presence of cardinal
PDS symptoms (postprandial fullness: P =0.05, early satiety:
P =0.02; Table 2 ) but not with the presence of cardinal EPS
symptoms (epigastric pain: P =0.18, epigastric burning: P =0.58;
Table 2 ).
not show signi cant correlations between neuronal response
characteristics and the number of eosinophils and mast cells
within the control or FD patient groups.
Correlation between neuronal impairment and clinical symptoms
in FD patients
We evaluated the association between neuronal impairment
in response to high-K
+
or ES in the submucous neurons of FD
patients and the prevalence of EPS or PDS symptoms (assessed
by using standardized questionnaires) ( 2,25,26 ). No correla-
tion was found between neuronal amplitudes and EPS (high-K
+
:
P =0.96 and P =0.51, ES: P =0.53 and P =0.91) or PDS (high-K
+
:
P =0.89 and P =1.00, ES: P =0.81 and P =0.64) symptoms (see
table 2 ).
Correlation between eosinophil/mast cell number and clinical
symptoms in FD patients
e association between the number of eosinophils and mast cells
and the prevalence of EPS or PDS symptoms ( 2,25,26 ) was also
evaluated in the submucous plexus of FD patients. Although no
correlation was found between the amount of mast cells and the
prevalence of EPS or PDS symptoms, intriguingly, the number
Eosinophils Mast cells
15
ΔF/F
0
(%)ΔF/F
0
(%)
ΔF/F
0
(%)ΔF/F
0
(%)
High-K
+
ES
20
25
10
5
0
15
20
25
10
5
0
1
2
3
4
5
1
2
3
4
5
0 50 100 150 200 250
0 50 100
Eosinophil cell number Mast cell number
FD patients Controls
150 200 250
200 400 500300100
200
400
500
300100
Figure 4 . Non-linear relationship between neuronal responses and the number of immune cells infi ltrated in the submucous plexus. ( a , b ) The graphs
show the Ca
2+
response amplitudes (Δ F/F
0
in %) elicited by a high-K
+
stimulus and numbers of eosinophils (left) and mast cells (right). ( c , d ) The graphs
display the data obtained after electrical stimulation (ES) and numbers of eosinophils (left) and mast cells (right). Black lines are non-linear fi ts through the
entire data set; the 95% confi dence interval for the fi tting parameters is indicated as a gray shade. Non-linear correlations (Spearman) were found in each
of the four comparisons. Individual data points were color coded (blue: controls, red: FD) post hoc to indicate the clinically defi ned group they belonged to.
Table 2 . Correlation between neuronal impairment and eosinophil/
mast cell number with prevalence of EPS or PDS symptoms
EPS ( n =10) PDS ( n =8)
Epigastric
pain
Epigastric
burning
Postprandial
fullness
Early
satiety
P value P value P value P value
High-K
+
0.96 0.51 0.89 1.00
ES 0.53 0.91 0.81 0.64
Eosinophils 0.18 0.58 0.05
a
0.02
a
Mast cells 0.79 1.00 0.61 0.87
EPS, epigastric pain syndrome; ES, electrical stimulation; PDS, postprandial
distress syndrome.
a
Spearman’s test.
The American Journal of GASTROENTEROLOGY www.amjgastro.com
1212
FUNCTIONAL GI DISORDERS
VOLUME 110
|
AUGUST 2015
Cirillo et al.
The subcellular localization of the neuronal HuCD and glial
S100 is different between FD patients and controls
In this study, we also assessed whether the HuCD staining dis-
played a typical cytosolic pattern or a distinct subcellular localiza-
tion ( 28 ) and found a more explicit nuclear staining in submucous
neurons of FD patients, compared with controls ( Figure 5a
and b ). To quantify this di erence, we used two methods. First,
we counted the neurons per ganglion having a strong nuclear
HuCD staining and found that this percentage was signi cantly
higher in FD patients compared with controls (66.5±5.3 vs.
41.0±4.2, P <0.01, Figure 5c ). To con rm these data, we also used
a second quanti cation method that calculates the mean HuCD
intensity ratio between the nucleus and the cytosol ( 28 ). Again,
this ratio was signi cantly higher in submucous neurons from
FD patients compared with controls (1.7±0.1 vs. 1.3±0.1, P <0.01,
Figure 5d ).
Along with the nuclear HuCD expression pattern, we found
the submucous glial cell labeling (using an antibody against S100)
to be more abundant in FD patients than in controls ( Figure 5a
and b ). We quanti ed this di erence by measuring the total
0
Controls
FD patients
20
Neurons with nuclear HuCD (%)
Mean intensity ratio
nucleus/cytosol (a.u.)
40
60
80
100
0
1
2
3
1000
500
Controls
FD patients
0
2000
Glia volume (mm
3
)/neuron
1500
2500
3000
**
**
**
Controls
HuCD S100
Merge
S100 volume
HuCD
FD patients
S100
Merge
S100 volume
Controls FD patients
Figure 5 . Neuronal and glial abnormalities in the submucous plexus of functional dyspepsia (FD) patients. The images represent neurons (HuCD, green)
and glial cells (S100, color code: magenta), as well as a merged image in controls ( a ) compared with FD patients ( b ). In controls, the staining of panneu-
ronal marker HuCD (green) is homogeneous in cytosol and nucleus (white arrows in a ), whereas in FD patients the intensity of the staining was higher in
the nucleus than in the cytosol (white arrows in b ). In controls, immunostaining shows that glial cells (color code: magenta) entangle neurons (green) and
form a well-defi ned mesh within the ganglion. In FD patients, the glial tangle (color code: magenta) around neurons (green) is more intricate, and single
glial cells are hard to identify. Bars = 20 μ m. ( c e ): the graphs show the percentage of submucous neurons with HuCD signal more intense in the nucleus
( c ), the mean intensity ratio nucleus/cytosol in submucous neurons, ( d ) and the ratio between the volume of S100 positive glial cells and the number of
neurons per ganglion ( e ) in controls (blue dots) compared with FD patients (red dots). ** P < 0.01.
© 2015 by the American College of Gastroenterology The American Journal of GASTROENTEROLOGY
1213
FUNCTIONAL GI DISORDERS
Submucous Defects in Functional Dyspepsia
ity. FD and IBS might be considered as a single disorder with
di erent manifestations. However, it is possible that in the two
disorders diverse cellular and molecular pathways are involved,
which may be responsible for di erent clinical manifestations.
Another di erence resides in the experimental approach used.
In our study, we directly assess the function of patient-speci c
neurons, whereas until now only reports were available that
have used neurons from experimental animals or human resec-
tion specimen to investigate the e ect of biopsy supernatants
( 14,15,18,19 ). us, at this moment a similar measurement in
biopsies from patients with IBS would be needed to directly
con rm or refute the fact that submucous neurons from IBS are
indeed hyperexcitable or whether soluble components from IBS
biopsies render neurons more excitable. Whether submucous
neurons themselves, which must have been exposed to these
components for a longer time, are still hyperexcitable is actually
still a matter of debate.
Although it is conceivable that, as a consequence of being
exposed to speci c stressors, maybe already for long time,
neurons might have become damaged, at the moment it is
not known what factors impair neuronal activation in FD
patients. One possible explanation might be found in a neuro–
immune interaction, which has been proposed to underlie
some of the pathophysiological expressions of FGIDs ( 30–34 ).
Indeed, Barbara et al. ( 30 ) demonstrated that mast cells are
in close proximity with nerve endings in the colonic mucosa
of patients with IBS. In line with this hypothesis, some recent
studies have shown immune activation and so-called ‘low-
grade in ammation’ in the duodenum of FD patients ( 8–10 ).
Here we add a new insight to this hypothesis, in that we meas-
ured a signi cant increase in the number of eosinophils and
mast cells, speci cally in the submucous plexus of FD patients.
It is plausible that the decreased neuronal Ca
2+
signaling in
submucous neurons of FD patients is due to the release of
noxious agents from surrounding cells or stroma. At this
moment, no conclusive proof is available, and further studies
will be needed to identify the time course, identity, and
concentration of the releasable factors that would result in a
dampened neuronal Ca
2+
response. An alternative thesis could
be that the neurons themselves are actually not a ected at all but
only become so during dissection, as immune mediators would
be released only from those biopsies containing a signi cant
amount of eosinophils and mast cells. However, the fact that
all dissections were performed at ice-cold temperatures miti-
gates any acute e ect of released biomolecules on neuronal Ca
2+
signaling.
To evaluate the symptomatic relevance of these  ndings, we
checked whether eosinophil and mast cell numbers correlated
with cardinal FD symptoms. We found a signi cant association
of submucous eosinophils with early satiety and postprandial
fullness, which are speci c symptoms of PDS. A similar correla-
tion in the duodenal mucosa of PDS patients has recently been
observed by Walker et al. ( 34 ). ey hypothesized that, through
mediator release and recruitment of mast cells, eosinophils
speci cally a ect neuronal and muscle cell functioning. We thus
volume of S100 positive cells and their processes relative to the
number of neurons per ganglion.  e total glial volume measured
in the submucous plexus of FD patients was signi cantly higher
than in controls (27224±2214.7 vs. 3659.8±272.3 μ m
3
, P <0.01). As
not all ganglia have the same size or numbers of neurons, we nor-
malized the glial volume to the number of neurons present in the
cubic selection and found this also to be signi cantly higher in FD
patients compared with controls (1993±382.4 vs. 362.1±139.8 μ m
3
,
respectively; P <0.01, Figure 5e ).
Neuronal VIP staining did not differ between FD patients and
controls
In an attempt to identify a certain subpopulation that might have
changed in the submucous plexus of FD patients, we performed
an immunohistochemical staining for VIP neurons, the most
prominent population in the submucous plexus ( 29 ). In all biop-
sies from controls and FD patients, we could detect VIP positivity
in nerve  bers and varicosities rather than in individual cell bod-
ies (data not shown).  e quanti cation of VIP staining did not
show any di erence between FD patients and controls in our set
of biopsies ( P =0.68).
DISCUSSION
e traditional assumption is that FGIDs are diseases
without organic features, hence the adjective ‘functional’ ( 1 ).
However, a rapidly increasing body of evidence shows that
subtle cellular and molecular alterations can be found in the
gastrointestinal tract of patients with FGIDs ( 4–10 ). In this
paper, we performed live imaging of neuronal activity, using
optical recording techniques in fresh nerve tissue (submucous
plexus) isolated from routine biopsies ( 21 ) to directly assess
whether nerve function is altered in FD patients. We also used
immunohistochemical analyses to assess possible changes in
the number of individual neurons, ganglia per biopsy, and neu-
rons per ganglia, which turned out to be comparable between
FD patients and controls.  e latter  nding helps explaining
why in classic pathological assessments cellular di erences are
le unnoticed.
e technology to perform live neuron recordings in routine
intestinal biopsies has only recently been established ( 21 ) and
here we demonstrate that it can be applied also in non-healthy
status. We found that there were no obvious di erences between
FD patients and controls in terms of how easily tissues could be
isolated and loaded with Ca
2+
indicator. However, we could show
that the amplitudes of neuronal responses in FD patients were
signi cantly lower compared with controls, both in response
to perfusion with high-K
+
and also to electrical stimulation.
Neuronal activity turned out to be similar when EPS and PDS
patients ( 2,25,26 ) were compared. Our data showing decreased
neuronal activity in FD patients appear in contrast to the  nd-
ings of other FGID (including IBS) studies, in which increased
neuronal activation has been suggested ( 14,15,18,19,30 ).  ere
are, however, a number of reasons why it is di cult to compare
these discordant  ndings, one of which may be disease speci c-
The American Journal of GASTROENTEROLOGY www.amjgastro.com
1214
FUNCTIONAL GI DISORDERS
VOLUME 110
|
AUGUST 2015
Cirillo et al.
hypothesized that a correlation might exist between increased
number of in ltrating eosinophils and neuronal impairment.
Although we did not  nd any correlation within the control
and the FD patient groups, we describe a signi cant negative
correlation between nerve activity and immune cell numbers
in the overall study population. As this analysis was performed
independently of clinical information and given the charac-
teristic distribution of data as revealed by post hoc labeling of
individual data points, our measured cellular parameters match
surprisingly well with the clinical subdivision between controls
and FD patients.  e non-linear correlation also establishes a
link between nerve function and the presence of immune cells
based on information obtained from live human samples. At
the moment it is not clear how acutely the neuronal function
is a ected, and it remains plausible that long-term e ects by
eosinophil-released mediators are needed to in uence neu-
ronal activity in FD patients. We can speculate that eosinophils,
a er migration into the submucous plexus, may a ect speci c
neuronal pathways responsible for early satiety and postpran-
dial fullness in PDS patients. As recently shown in an elegant
series of animal experiments, immune cells also contribute to
organizing motility by interacting with enteric nerves even
in a non-in amed condition ( 27 ).  e correlation between
cellular/molecular  ndings with clinical symptoms in FD, as
well as in other FGIDs has been evaluated in several studies
( 8–10,30,34 ), but it is still controversial. It may well be that,
although underlying cellular defects are common, symptom
reporting is much more related to individual di erences in
perception and may su er from linguistic limitations in symp-
tom description repertoires, as well as modulation by many
other factors (social, psychological) known to be involved ( 35 ).
Further investigation into these other aspects is thus needed to
narrow the distance between lab bench measurements and clini-
cal evaluations.
Together with neuronal functionality, we analyzed the mor-
phology of submucous neurons in FD patients by using speci c
markers. First of all, immunohistochemical labeling revealed
that the neuronal marker HuCD ( 28,36 ) localized di erently in
neurons of FD patients compared with controls. We and others
( 28,36–38 ) have shown that mouse enteric neurons under path-
ological circumstances, which could be induced by mechanical
damage and hypoxia, display a higher nuclear HuCD appear-
ance. Although at present there is no conclusive explanation
for the altered HuCD expression in submucous neurons of FD
patients, it may be taken along as another measurable cellular
change. In order to identify a speci c neuronal subpopulation
that might be a ected in FD patients, we analyzed VIP stain-
ing, which labels a major class of neurons and nerve  bers in
the submucous plexus ( 29 ). In our set of biopsies, we could not
detect di erences in VIP staining, which labeled nerve  bers
rather than nerve cell bodies both in FD patients and controls.
However, we cannot exclude that a speci c neuronal subset
might be more a ected compared with other subpopulations
during FD.
As gut function is the result of a combined and fine inter-
play between all enteric nervous system components, we also
analyzed glial morphology in the submucous plexus of FD
patients. Glial cells are tightly packed with neurons to pro-
vide structural and functional support to them. Interestingly,
we measured the abundant presence of the glial marker S100
protein around the submucous neurons of FD patients. It is
widely described that, when triggered, glial cells react and
overexpress glial-derived molecules, a phenomenon called
reactive gliosis and described in many intestinal diseases char-
acterized by inflammation, like celiac disease, ulcerative coli-
tis, and other inflammatory neuropathies ( 39–43 ). Although
it is well established that glial cells are key players in several
gut diseases ( 44,45 ), this is the first study that directly meas-
ured neuronal impairment and gliosis in (FD) patients. On the
basis of our findings, we can speculate that, in the submucous
plexus of FD patients, inflammatory mediators released by
eosinophils and mast cells trigger glial cells to in turn release
factors (cytokines, nitric oxide, nerve growth factor) that affect
neuronal functioning ( 46,47 ).
In summary, we demonstrated impairment of neuronal signal-
ing and altered expression patterns of neuronal and glial markers
in submucous plexus of FD patients.  e structural and func-
tional abnormalities identi ed in this study are indicative of a
subtle type of enteric neuro and gliopathy present in FD patients.
Here we show that such abnormalities can be directly monitored
in easy-accessible tissues such as duodenal biopsies by optical
techniques.
ACKNOWLEDGMENTS
We thank the members of LENS for their critical comments and
skilled technical assistance. We also thank Maura Corsetti, Ingrid
Demedts, Florencia Carbone, Natália Pessoa Rocha, and Lukas Van
Oudenhove (TARGID) for the insightful comments and useful
discussions. C.C. and H.V. are post-doctoral fellows and A.-S.D.
a doctoral fellow of the Fonds voor Wetenschappelijk Onderzoek
(FWO, Belgium).
CONFLICT OF INTEREST
Guarantor of the article: Pieter Vanden Berghe, MSc, PhD.
Speci c author contributions : Study concept and design, acquisi-
tion of data, analysis and interpretation of data, dra ing and
editing of the manuscript, and statistical analysis: C.C. and P.V.B;
provision of biopsy material: T.B. and J.T.; enrollment of patients
for the study: T.B. and H.V.; acquisition and analysis of the data:
A.-S.D.; critical revision of the manuscript: T.B., A.-S.D., H.V., and
J.T.; P.V.B. wrote the analysis so ware; and J.T. and P.V.B. obtained
funding.
Financial support: is work was funded by BOF, University of
Leuven (Methusalem Jan Tack; OT/STRT1 Pieter Vanden Berghe)
and FWO (G0889.11; G.0A44.13). Confocal recordings were made
on the equipment of the Cell Imaging Core (University of Leuven)
supported by Hercules foundation grants to P.V.B.
Potential competing interests: None.
© 2015 by the American College of Gastroenterology The American Journal of GASTROENTEROLOGY
1215
FUNCTIONAL GI DISORDERS
Submucous Defects in Functional Dyspepsia
19. Krueger D , Michel K , Allam S et al. E ect of hyoscine butylbromide
(Buscopan) on cholinergic pathways in the human intestine . Neurogastro-
enterol Motil 2013 ; 25 : e530 – 9 .
20. Miwa H , Watari J , Fukui H et al. Current understanding of pathogenesis of
functional dyspepsia . J Gastroenterol Hepatol 2011 ; 26 : 53 – 60 .
21. Cirillo C , Tack J , Vanden Berghe P . Nerve activity recordings in routine
human intestinal biopsies . Gut 2013 ; 62 : 227 – 35 .
22. Boesmans W , Martens MA , Weltens N et al. Imaging neuron-glia inter-
actions in the enteric nervous system . Front Cell Neurosci 2013 ; 7 : 183 .
23. Lebouvier T , Coron E , Chaumette T et al. Routine colonic biopsies as a new
tool to study the enteric nervous system in living patients . Neurogastro-
enterol Motil 2010 ; 22 : e11 – 4 .
24. Rotondano G . Reducing complications in upper gastrointestinal endoscopy .
Expert Rev Gastroenterol Hepatol 2012 ; 6 : 271 – 90 .
25. Tack J , Talley NJ . Gastroduodenal disorders . Am J Gastroenterol 2010 ; 105 : 757 – 63 .
26. Barlow WJ , Orlando RC . e pathogenesis of heartburn in nonerosive
re ux disease: a unifying hypothesis . Gastroenterology 2005 ; 128 : 771 – 8 .
27. Muller PA , Koscsó B , Rajani GM et al. Crosstalk between muscularis macrophag-
es and enteric neurons regulates gastrointestinal motility . Cell 2014 ; 158 : 300 – 13 .
28. Desmet AS , Cirillo C , Vanden Berghe P . Distinct subcellular localization
of the neuronal marker HuC/D reveals hypoxia-induced damage in enteric
neurons . Neurogastroenterol Motil 2014 ; 26 : 1131 – 43 .
29. Costa M , Furness JB , Gibbins IL et al. Colocalization of VIP with other
neuropeptides and neurotransmitters in the autonomic nervous system .
Ann NY Acad Sci 1988 ; 527 : 103 – 9 .
30. Barbara G , Stanghellini V , De Giorgio R et al. Activated mast cells in proxi-
mity to colonic nerves correlate with abdominal pain in irritable bowel
syndrome . Gastroenterology 2004 ; 126 : 693 – 702 .
31. Wouters MM , Boeckxstaens GE . Neuroimmune mechanisms in functional
bowel disorders . Neth J Med 2011 ; 69 : 55 – 61 .
32. Collins SM , Barbara G . East meets West: infection, nerves, and mast cells in
the irritable bowel syndrome . Gut 2004 ; 53 : 1068 – 9 .
33. Walker MM , Salehian SS , Murray CE et al. Implications of eosinophilia in
the normal duodenal biopsy–an association with allergy and functional
dyspepsia . Aliment Pharmacol er 2010 ; 31 : 1229 – 36 .
34. Walker MM , Aggarwal KR , Shim LS et al. Duodenal eosinophilia and early
satiety in functional dyspepsia: con rmation of a positive association in an
Australian cohort . J Gastroenterol Hepatol 2014 ; 29 : 474 – 9 .
35. Van Oudenhove L , Aziz Q . e role of psychosocial factors and psychiatric dis-
orders in functional dyspepsia . Nat Rev Gastroenterol Hepatol 2013 ; 10 : 158 – 67 .
36. Phillips RJ , Hargrave SL , Rhodes BS et al. Q u a n t i cation of neurons in the
myenteric plexus: an evaluation of putative pan-neuronal markers . J Neuro-
sci Met 2004 ; 133 : 99 – 107 .
37. Phillips RJ , Kie er EJ , Powley TL . Loss of glia and neurons in the myenteric
plexus of the aged Fischer 344 rat . Anat Embryol 2004 ; 209 : 19 – 30 .
38. acker M , Rivera LR , Cho HJ et al. e relationship between glial distor-
tion and neuronal changes following intestinal ischemia and reperfusion .
Neurogastroenterol Motil 2011 ; 23 : e500 – 9 .
39. Esposito G , Cirillo C , Sarnelli G et al. Enteric glial-derived s100b protein
stimulates nitric oxide production in celiac disease . Gastroenterology
2007 ; 133 : 918 – 25 .
40. Cirillo C , Sarnelli G , Esposito G et al. Increased mucosal nitric oxide pro-
duction in ulcerative colitis is mediated in part by the enteroglial-derived
S100B protein . Neurogastroenterol Motil 2009 ; 21 : 1209 – e112 .
41. Cirillo C , Sarnelli G , Esposito G et al. S100B protein in the gut: the evidence
for enteroglial-sustained intestinal in ammation . World J Gastroenterol
2011 ; 17 : 1261 – 6 .
42. Bernardini N , Segnani C , Ippolito C et al. Immunohistochemical analysis
of myenteric ganglia and interstitial cells of Cajal in ulcerative colitis . J Cell
Mol Med 2012 ; 16 : 318 – 27 .
43. Fagbemi AO , Torrente F , Puleston J et al. Enteric neural disruption in ne-
crotizing enterocolitis occurs in association with myenteric glial cell CCL20
expression . J Pediatr Gastroenterol Nutr 2013 ; 57 : 788 – 93 .
44. Nasser Y , Fernandez E , Keenan CM et al. Role of enteric glia in intestinal
physiology: e ects of the gliotoxin 
uorocitrate on motor and secretory
function . Am J Physiol Gastrointest Liver Physiol 2006 ; 291 : G912 – 27 .
45. Vasina V , Barbara G , Talamonti L et al. Enteric neuroplasticity evoked by
in ammation . Auton Neurosci 2006 ; 126-127 : 264 – 72 .
46. Murakami M , Ohta T , Ito S . Lipopolysaccharides enhance the action of
bradykinin in enteric neurons via secretion of interleukin-1beta from
enteric glial cells . J Neurosci Res 2009 ; 87 : 2095 – 104 .
47. von Boyen GB , Steinkamp M , Reinshagen M et al. Nerve growth factor
secretion in cultured enteric glia cells is modulated by proin ammatory
cytokines . J Neuroendocrinol 2006 ; 18 : 820 – 5 .
Study Highlights
WHAT IS CURRENT KNOWLEDGE
Functional dyspepsia (FD) is a common gastrointestinal
disorder without clear etiology and pathophysiology.
Recent evidence points the attention to cellular and
molecular abnormalities.
The role of enteric nerves in the pathophysiology of FD has
been only indirectly demonstrated.
WHAT IS NEW HERE
Submucous neuronal function is impaired in FD.
Ganglionic architecture, as revealed by glial and neuronal
markers, shows crucial abnormalities in FD.
Altered neuronal function correlates with eosinophil and
mast cell infi ltration.
REFERENCES
1. Knowles CH , Lindberg G , Panza E et al. New perspectives in the diagnosis
and management of enteric neuropathies . Nat Rev Gastroenterol Hepatol
2013 ; 10 : 206 – 18 .
2 . T a c k J , T a l l e y N J , C a m i l l e r i M et al. Functional gastroduodenal disorders .
Gastroenterology 2006 ; 130 : 1466 – 79 .
3. Talley NJ . Functional gastrointestinal disorders as a public health problem .
Neurogastroenterol Motil 2008 ; 20 ( Suppl 1 ): 121 – 9 .
4 . P i t t o c k S J , L e n n o n V A , D e g e C L et al. Neural autoantibody evaluation
in functional gastrointestinal disorders: a population-based case-control
study . Dig Dis Sci 2011 ; 56 : 1452 – 9 .
5. Larsson MH , Simrén M , omas EA et al. Elevated motility-related trans-
mucosal potential di erence in the upper small intestine in the irritable
bowel syndrome . Neurogastroenterol Motil 2007 ; 19 : 812 – 20 .
6. Bassotti G , Villanacci V , Creţoiu D et al. Cellular and molecular basis of
chronic constipation: taking the functional/idiopathic label out . World J
Gastroenterol 2013 ; 19 : 4099 – 105 .
7. El-Salhy M , Gundersen D , Ostgaard H et al. Low densities of serotonin and
peptide YY cells in the colon of patients with irritable bowel syndrome .
Dig Dis Sci 2012 ; 57 : 873 – 8 .
8 . V a n h e e l H , V i c a r i o M , V a n u y t s e l T et al. Impaired duodenal mucosal
integrity and low-grade in ammation in functional dyspepsia . Gut
2014 ; 63 : 262 – 71 .
9. Walker MM , Talley NJ . Functional gastrointestinal disorders and the poten-
tial role of eosinophils . Gastroenterol Clin North Am 2008 ; 37 : 383 – 95 .
10. Walker MM , Talley NJ , Prabhakar M et al. Duodenal mastocytosis, eosino-
philia and intraepithelial lymphocytosis as possible disease markers in the
irritable bowel syndrome and functional dyspepsia . Aliment Pharmacol
er 2009 ; 29 : 765 – 73 .
11. Furness JB . e enteric nervous system: normal functions and enteric
neuropathies . Neurogastroenterol Motil 2008 ; 20 : 32 – 8 .
12. Furness JB . e enteric nervous system and neurogastroenterology . Nat Rev
Gastroenterol Hepatol 2012 ; 9 : 286 – 94 .
13. Obermayr F , Hotta R , Enomoto H et al. Development and developmental
disorders of the enteric nervous system . Nat Rev Gastroenterol Hepatol
2013 ; 10 : 43 – 57 .
14. Barbara G , Wang B , Stanghellini V et al. Mast cell-dependent excitation of
visceral-nociceptive sensory neurons in irritable bowel syndrome . Gastro-
enterology 2007 ; 132 : 26 – 37 .
15. Balestra B , Vicini R , Cremon C et al. Colonic mucosal mediators from
patients with irritable bowel syndrome excite enteric cholinergic motor
neurons . Neurogastroenterol Motil 2012 ; 24 : 1118 – e570 .
16. Grover M , Camilleri M . E ects on gastrointestinal functions and symptoms
of serotonergic psychoactive agents used in functional gastrointestinal
diseases . J Gastroenterol 2013 ; 48 : 177 – 81 .
17. Tack J , Demedts I , Dehondt G et al. Clinical and pathophysiological
characteristics of acute-onset functional dyspepsia . Gastroenterology
2002 ; 122 : 1738 – 47 .
18. Buhner S , Li Q , Vignali S et al. Activation of human enteric neurons by
supernatants of colonic biopsy specimens from patients with irritable bowel
syndrome . Gastroenterology 2009 ; 137 : 1425 – 34 .
... 10,11 This inflammation may lead to decreased mucosal integrity and abnormal smooth muscle contractions. 12,13 Previous research from our group has shown that a combination of antispasmodic and anxiolytic drugs (clidinium bromide/chlordiazepoxide) improves symptoms and quality of life in FD patients refractory to PPIs. 14 However, the effect of antispasmodic monotherapy as an add-on treatment to PPIs in FD remains undefined. ...
... Our hypothesis of interest was duodenal low-grade inflammation causing abnormal contraction of intestinal smooth muscles resulting in dyspeptic symptoms. [10][11][12][13] Our results suggest that the modulation of abnormal intestinal contraction may not be beneficial for all patients with FD. However, subtype analysis revealed improvement in early satiety, a common symptom of PDS. ...
Article
Full-text available
Background and Aim Functional dyspepsia (FD) remains a therapeutic challenge, and the efficacy of antispasmodic agents as adjunctive therapy is not well established. This study aimed to evaluate the efficacy and safety of pinaverium bromide added to omeprazole in treating refractory FD. Methods We conducted a randomized, placebo‐controlled trial in patients with refractory dyspepsia. Participants were randomly assigned to receive pinaverium (50 mg, 3 times/day, n = 36) or placebo ( n = 36) in addition to omeprazole for 8 weeks. The primary endpoint was the responder rate for adequate relief. Secondary outcomes included the Global Overall Symptom Scale (GOSS), quality of life, and safety profile. Results No statistically significant differences were observed in the adequate relief response rate between the pinaverium bromide and control group at week 2 (58.3% vs. 62.9%, P = 0.697), week 4 (62.9% vs. 78.1%, P = 0.173), week 6 (64.7% vs. 75.0%, P = 0.363), and week 8 (64.7% vs. 75.0%, P = 0.363). Additionally, there were no significant differences observed in the decline of symptom score between the two groups at week 4 (8.4 ± 7.6 vs. 7.7 ± 7.1, P = 0.702) and week 8 (10.9 ± 8.2 vs. 8.4 ± 7.2, P = 0.196). Similarly, there were no significant differences in terms of quality of life between the two groups. Adverse event rates were also comparable between the two groups. Conclusion Pinaverium bromide was found to be safe in the treatment of refractory dyspepsia, but it did not demonstrate a significant benefit in improving symptoms.
... Our results are consistent with those of previous studies, in which the increase in eosinophils and mast cells was significantly increased in the FD group compared to the controls, and an increase in duodenal Streptococcus. [21][22][23] The relationship between duodenal mucosal inflammation and FD symptoms, as demonstrated in previous studies, served as a means to validate the reliability of the samples obtained from the study cohort. The two main new findings of our research are as follows: the identification of previously unidentified novel microbial genera associated with FD in stool samples, and the relationship between dietary micronutrition and FD. ...
Article
Background/aims: : Functional dyspepsia (FD) has long been regarded as a syndrome because its pathophysiology is multifactorial. However, recent reports have provided evidence that changes in the duodenal ecosystem may be the key. This study aimed to identify several gastrointestinal factors and biomarkers associated with FD, specifically changes in the duodenal ecosystem that may be key to understanding its pathophysiology. Methods: : In this case-control study, 28 participants (12 with FD and 16 healthy control individuals) were assessed for dietary nutrients, gastrointestinal symptom severity, immunological status of the duodenal mucosa, and microbiome composition from oral, duodenal, and fecal samples. Integrated data were analyzed using immunohistochemistry, real-time polymerase chain reaction, 16S rRNA sequencing, and network analysis. Results: : Duodenal mucosal inflammation and impaired expression of tight junction proteins were confirmed in patients with FD. The relative abundance of duodenal Streptococcus (p=0.014) and reductions in stool Butyricicoccus (p=0.047) were confirmed. These changes in the gut microbiota were both correlated with symptom severity. Changes in dietary micronutrients, such as higher intake of valine, were associated with improved intestinal barrier function and microbiota. Conclusions: : This study emphasizes the relationships among dietary nutrition, oral and gut microbiota, symptoms of FD, impaired function of the duodenal barrier, and inflammation. Assessing low-grade inflammation or increased permeability in the duodenal mucosa, along with changes in the abundance of stool Butyricicoccus, is anticipated to serve as effective biomarkers for enhancing the objectivity of FD diagnosis and monitoring.
... 55 In support of eosinophils playing a pathogenic role in FD, alterations in duodenal neuronal structure and function have been documented in FD versus controls, and increased small intestinal permeability has been observed; these alterations correlate with the microscopic inflammation present. 39,56,57 Rumination syndrome characterized by effortless regurgitation is now recognized to be a relatively common DGBI that significantly overlaps with FD. In a US population-based study, the prevalence of rumination syndrome and FD was surprisingly similar (5.8% and 7.1%, respectively) and these disorders overlapped four-times more often than expected by chance. ...
Article
Full-text available
Functional dyspepsia (FD) is a highly prevalent disorder. Upper endoscopy is normal, and according to the Rome IV criteria, there is no established pathology. Data accumulated over the last 15 years has challenged the notion FD is free of relevant pathology, and in particular, increased duodenal eosinophils have been observed. Intestinal eosinophils play important roles in microbial defence, immune regulation, tissue regeneration and remodelling, and maintaining tissue homeostasis and metabolism; degranulation of eosinophils releases toxic granule products (e.g., major basic protein, eosinophil-derived neurotoxin) which can damage nerves. Normal cut-offs for eosinophil infiltration into the duodenum histologically are less than five eosinophils per high power field (<25 per five high power fields). In clinical practice there is evidence that pathologically increased intestinal eosinophils may often be overlooked. In a meta-analysis duodenal eosinophils were significantly increased in FD although there was substantial heterogeneity; degranulation of duodenal eosinophils was also significantly higher in FD without significant heterogeneity. In addition, increased duodenal permeability, systemic immune activation, and an altered mucosa-associated duodenal microbiome have been identified that may help explain why symptoms arise, often occur after food with exposure to food antigens, and typically fluctuate. Several potentially reversible risk factors for FD have now been identified. We evaluate the current evidence linking duodenal microinflammation and immune activation with FD and disorders of gut–brain interactions that overlap with FD. We propose a two-hit disease model for eosinophilic functional dyspepsia (EoFD) with management implications.
... Исследование, проведенное С. Cirillo, задокументировало инфильтрацию подслизистого слоя тучными клетками [15]. В другом исследовании, проведенном F. Tanaka [16], у пациентов с ФД экспрессия НФГК была увеличена и дополнительно коррелировала с симптомами. ...
Article
Functional dyspepsia, affecting up to 20% of individuals worldwide, remains both a cause of decreased activity of patients’ daily life and an obvious economic burden due to healthcare costs. Despite extensive research, the etiology of dyspepsia is unknown in most patients. Intestinal motility dysfunction has long been considered the major culprit, but recent studies suggest that immune pathophysiological and molecular effects in the duodenum are far more likely predisposing factors. Eosinophilia and an increase in mast cells in both the duodenum and gastric mucosa are identified in most patients with this disease. More and more data on the significant role of impaired paracellular permeability of the intestinal mucosa are now available. It is associated with subclinical inflammation in the submucosal layer in patients with functional dyspepsia. This explains the poor effectiveness of the treatments taken. The evidence from practice suggests that symptoms persist or return after eradication therapy in most patients. Proton pump inhibitors and antidepressants do not ease postprandial distress syndrome. Montelukast and cromolyn therapy has been proposed, but this approach is not yet widely popular. Therefore, there is an obvious need in finding other therapeutic approaches. One of them is the increased use of prokinetics, the most recent of which is acotiamide. Its mechanism of action is similar to that of prior generation prokinetics (inhibition of acetylcholinesterase activity), but is distinguished by the absence of impact on dopaminergy, due to which the drug has far fewer side effects. In addition, its effect on the production of ghrelin, which physiological role is being actively studied, is discussed.
Article
Mast cells have been implicated in abdominal pain-associated disorders of gut-brain interaction, such as functional dyspepsia. As such, ketotifen, a second-generation antihistamine and mast cell stabilizer, could represent a viable treatment option in these conditions. The primary aim of the current pilot study was to assess clinical response to ketotifen and assess pharmacokinetics in youth with functional dyspepsia. We conducted a pilot randomized, double-blind, placebo-controlled, cross-over trial of ketotifen in 11 youth with functional dyspepsia and duodenal mucosal eosinophilia with 4 weeks of active treatment at a dose of 1 mg twice daily. Global clinical response was graded on a 5-point Likert Scale. A single plasma sample was obtained at steady state for pharmacokinetic analysis. Ketotifen was not superior to placebo with regard to global clinical response. Only 18% of patients demonstrated a complete or near-complete clinical response. The estimated half-life was 3.3 h. While ketotifen was not superior to placebo, this study highlights several important challenges for developing drug trials for youth with chronic abdominal pain. Recommendations are made for designing a larger treatment trial for ketotifen in this patient group. This study was registered at ClinicalTrials.gov: NCT02484248
Article
Full-text available
Background and purpose Functional dyspepsia (FD) is a prevalent global disorder of the upper digestive tract characterized by functional impairment. It often coexists with anxiety/depression, significantly impairing occupational productivity and overall quality of life. This study aimed to identify emerging patterns and prominent themes within FD and anxiety/depression research through bibliometric analysis to help explore new innovative avenues for investigating this type of FD. Methods A comprehensive review of literature encompassing FD and anxiety/depression was conducted using the Science Citation Index Extension of the Web of Science Core Collection from 2003 to 2023. Information extracted comprised “Full Record and Cited References.” Bibliometric analysis of relevant publications, including country, institution, author, journal, citations, and keywords, was conducted using CiteSpace, VOSviewer, and Bibliometrix package in R and Excel. Results Studies related to FD and anxiety/depression have demonstrated an ascending trajectory since 2003. Our bibliometric analysis identified 338 studies published by 2023. NEUROGASTROENTEROLOGY AND MOTILITY emerged as the most prolific journal, while GASTROENTEROLOGY retained pre-eminence within the top 10 published journals. China emerged as the most prolific country, with two institutions within the top 10 in terms of volume of publications. The Mayo Clinic stood as the foremost institution in terms of publication volume, with the Chengdu University of Traditional Chinese Medicine exhibiting robust collaborative engagement. Eminent author influence was attributed to Talley NJ of Newcastle University, Australia. Clusters of extensively cited papers and prevalent keywords delineate the status and trend of FD and anxiety/depression research. This encompasses FD, anxiety, depression, sleep disorders, and functional gastrointestinal disorders. Furthermore, the timeline view map or trend-term analysis suggested that duodenal low-grade inflammation (“duodenal eosinophilia” and “mast cells”) might be a new concern associated with FD and anxiety/depression. Conclusion Employing bibliometric analysis, this study revealed prevalent focal areas and new trends within FD and anxiety/depression research. These insights serve as valuable guidance for scholars seeking to delve into new research avenues.
Article
Background: Exosomes are applied as biomarkers in several diseases according to their disease-specific profiles. However, the exosomes effects in functional dyspepsia (FD) are still fragmentary. Here we examined the role of Eosinophil and mast cell derived-exosomes in FD progression. Methods: Fifty FD subjects and age- and sex-matched healthy controls were included in this retrospective cohort study. Duodenal mucosa and gastric juice were collected to analyze molecular difference. Eosinophil and mast cell were evaluated by immunofluorescence and microarray was subjected to examine the expression levels of NEAT1, miR-211-5p, and glial cell line-derived neurotrophic factor (GDNF), which were subsequently were tested by quantitative reverse transcription PCR (RT-qPCR) validation cohorts. CCK-8 assays, and wound healing assays were used to evaluate integrity of intestinal mucosal barrier in vitro. Rats' weights and gastric emptying rates were used as evaluation of FD severity in vivo. Results: Eosinophil and mast cell were enriched and secreted more exosomes in duodenal mucosa of FD patients. We identified differential lncRNAs that were consistently and significantly up regulated in FD cases. Of these, NEAT1 was further validated by RT-qPCR and had closely relationship with GDNF. MiR-211-5p level was found to be reduced in FD and negatively related with NEAT1 and GDNF. Furthermore, NEAT1and GDNF relived FD while miR-211-5p made symptoms worse. The NEAT1/miR-211-5p/GDNF axis had a good predictive ability for FD. Conclusions: The NEAT1/miR-211-5p/GDNF could be a potential FD biomarker.
Article
In the body, nerve tissue is not only present in the central nervous system, but also in the periphery. The enteric nervous system (ENS) is a highly organized intrinsic network of neurons and glial cells grouped to form interconnected ganglia. Glial cells in the ENS are a fascinating cell population: their neurotrophic role is well established, as well as their plasticity in specific circumstances. Gene expression profiling studies indicate that ENS glia retain neurogenic potential. The identification of neurogenic glial subtype(s) and the molecular basis of glia-derived neurogenesis may have profound biological and clinical implications. In this review, we discuss the potential of using gene-editing for ENS glia and cell transplantation as therapies for enteric neuropathies. Glia in the ENS: target or tool for nerve tissue repair?
Article
Ethnopharmacological relevance: Functional dyspepsia (FD) is a disorder caused by abnormal gut-brain axis regulation and is highly prevalent in China. Cynanchum auriculatum (CA) is often used to treat FD in the ethnic minority areas of Guizhou. Although several CA-based products are currently available in the market, it is unclear which components of CA are efficacious and what their oral absorption mechanism is. Aim of the study: This study aimed to screen anti-FD components of CA based on the spectrum-effect relationship. In addition, the study evaluated the intestinal absorption mechanism of these components using transporter inhibitors. Materials and methods: The fingerprinting of compounds from CA extract and plasma after oral administration was conducted using ultra-high-performance liquid chromatography quadrupole-time-of-flight tandem mass spectrometry (UHPLC-Q-TOF-MS). The intestinal contractile parameters were then measured in vitro using the BL-420F Biofunctional Experiment System. Multivariate statistical analysis of the result of spectrum-effect relationship assessment was used to elucidate the correlation between prominent peaks of CA-containing plasma and intestinal contractile activity. The effect of ATP-binding cassette (ABC) transporter inhibitors, such as the P-gp inhibitor verapamil, the MRR inhibitor indomethacin, and the BCRP inhibitor Ko143, on the directional transport of the predicted active ingredients was assessed in vivo. Results: Twenty chromatographic peaks were identified in the CA extract. Of these, three were C21 steroids, four were organic acids, and one was a coumarin, and acetophenone by comparing with reference compounds. Additionally, it is discovered that there are totally 39 migratory components in CA-containing plasma, which was found to significantly promote the contractility of the isolated duodenum. Moreover, multivariate analysis of the spectrum-effect relationship demonstrated that 16 characteristic peaks (3, 6, 8, 10, 11, 13, 14, 18, 21, m1-m4, m7, m15, and m24) in CA-containing plasma were significantly associated with the anti-FD effect. These compounds included seven prototype compounds, i.e., cynanoneside A, syringic acid, deacylmetaplexigenin, ferulic acid, scopoletin, baishouwubenzophenone, and qingyangshengenin. The inhibition of ABC transporters demonstrated that the inhibitors verapamil and Ko143 significantly increased (P < 0.05) the uptake of scopoletin and qingyangshengenin. Thus, these compounds may be substrates for P-gp and BCRP. Conclusions: The potential anti-FD components of CA and the effect of ABC transporter inhibitors on these active components were preliminarily clarified. These findings lay a foundation for subsequent in vivo studies.
Article
Background: Functional dyspepsia (FD) is a multifactorial disorder. Helicobacter pylori (H. pylori)-related dyspepsia (HpD) may be considered a separate entity. Duodenal eosinophilia is a potential pathogenic mechanism in FD. However, the impact of duodenal eosinophilia and host genetic polymorphism of innate and pro-inflammatory cascade, nucleotide-binding oligomerization domain 1 (NOD-1), and interleukin-1 beta (IL-1β) in HpD was not explored. Aim: To evaluate the association of NOD1-796G>A and IL-1B-511C>T gene variants and low-grade duodenal eosinophilia in HpD. Methods: A multicenter cross-sectional study was conducted. A total of 253 patients who met Rome-IV criteria were selected before upper endoscopy and 98 patients were included after unremarkable upper endoscopy and positive H. pylori in gastric biopsies were assessed. Clinical parameters, H. pylori cagA and duodenal histology, were evaluated. Results: Sixty-four (65%) patients had epigastric pain syndrome (EPS), 24 (25%) postprandial distress syndrome (PDS), and 10 (10%) EPS/PDS overlap. FD subtypes were not associated with NOD1-796G>A and IL-1B-511C>T gene variants. Low-grade duodenal eosinophilia was significantly increased in NOD1-796 GG versus single A-allele, but not in IL-1B-511 single T-allele or CC-allele. This association is dependent of cagA infection, since harboring cagA strain was significantly associated with low-grade duodenal eosinophilia with isolated variants NOD1-796 GG and IL-1B-511 single T-allele, but not without cagA. When we performed combined polymorphism analysis with NOD1-796 GG/IL-1B-511 single T-allele, a synergistic effect on low-grade duodenal eosinophilia was found between these two loci irrespective of cagA strain status in HpD. Conclusion: Our findings suggest that low-grade duodenal eosinophilia is significantly associated with NOD1-796 GG allele specially in cagA strain and with allelic combination NOD1-796 GG/IL-1B-511 single T-allele independent of cagA strain infection in HpD patients.
Article
Full-text available
Intestinal peristalsis is a dynamic physiologic process influenced by dietary and microbial changes. It is tightly regulated by complex cellular interactions; however, our understanding of these controls is incomplete. A distinct population of macrophages is distributed in the intestinal muscularis externa. We demonstrate that, in the steady state, muscularis macrophages regulate peristaltic activity of the colon. They change the pattern of smooth muscle contractions by secreting bone morphogenetic protein 2 (BMP2), which activates BMP receptor (BMPR) expressed by enteric neurons. Enteric neurons, in turn, secrete colony stimulatory factor 1 (CSF1), a growth factor required for macrophage development. Finally, stimuli from microbial commensals regulate BMP2 expression by macrophages and CSF1 expression by enteric neurons. Our findings identify a plastic, microbiota-driven crosstalk between muscularis macrophages and enteric neurons that controls gastrointestinal motility. PAPERFLICK:
Article
Full-text available
The enteric nervous system (ENS) is a network of neurons and glia within the wall of the gastrointestinal tract that is able to control many aspects of digestive function independently from the central nervous system. Enteric glial cells share several features with astrocytes and are closely associated with enteric neurons and their processes both within enteric ganglia, and along interconnecting fiber bundles. Similar to other parts of the nervous system, there is communication between enteric neurons and glia; enteric glial cells can detect neuronal activity and have the machinery to intermediate neurotransmission. However, due to the close contact between these two cell types and the particular characteristics of the gut wall, the recording of enteric glial cell activity in live imaging experiments, especially in the context of their interaction with neurons, is not straightforward. Most studies have used calcium imaging approaches to examine enteric glial cell activity but in many cases, it is difficult to distinguish whether observed transients arise from glial cells, or neuronal processes or varicosities in their vicinity. In this technical report, we describe a number of approaches to unravel the complex neuron-glia crosstalk in the ENS, focusing on the challenges and possibilities of live microscopic imaging in both animal models and human tissue samples.
Article
Full-text available
In recent years, the improvement of technology and the increase in knowledge have shifted several strongly held paradigms. This is particularly true in gastroenterology, and specifically in the field of the so-called "functional" or "idiopathic" disease, where conditions thought for decades to be based mainly on alterations of visceral perception or aberrant psychosomatic mechanisms have, in fact, be reconducted to an organic basis (or, at the very least, have shown one or more demonstrable abnormalities). This is particularly true, for instance, for irritable bowel syndrome, the prototype entity of "functional" gastrointestinal disorders, where low-grade inflammation of both mucosa and myenteric plexus has been repeatedly demonstrated. Thus, researchers have also investigated other functional/idiopathic gastrointestinal disorders, and found that some organic ground is present, such as abnormal neurotransmission and myenteric plexitis in esophageal achalasia and mucosal immune activation and mild eosinophilia in functional dyspepsia. Here we show evidence, based on our own and other authors' work, that chronic constipation has several abnormalities reconductable to alterations in the enteric nervous system, abnormalities mainly characterized by a constant decrease of enteric glial cells and interstitial cells of Cajal (and, sometimes, of enteric neurons). Thus, we feel that (at least some forms of) chronic constipation should no more be considered as a functional/idiopathic gastrointestinal disorder, but instead as a true enteric neuropathic abnormality.
Article
Full-text available
Objective: Functional dyspepsia (FD) is an extremely common functional gastrointestinal disorder, the pathophysiology of which is poorly understood. We hypothesised that impaired intestinal barrier function is involved in the onset and persistence of this disorder by inducing low-grade inflammation. Therefore, our aim was to evaluate duodenal mucosal integrity and low-grade inflammation in patients with FD. Design: Duodenal biopsy specimens were obtained from 15 patients with FD fulfilling the Rome III criteria and 15 age- and gender-matched healthy volunteers. Transepithelial electrical resistance (TEER) and paracellular permeability were measured in Ussing chambers. Expression of cell-to-cell adhesion proteins was evaluated by real-time PCR, western blot and/or immunofluorescence. Numbers of mast cells, eosinophils and intraepithelial lymphocytes were assessed by immunohistochemistry. Results: Patients with FD displayed lower TEER and increased paracellular passage compared with healthy controls, which is indicative of impaired mucosal integrity. In addition, abnormal expression of cell-to-cell adhesion proteins at the level of tight junctions, adherens junctions and desmosomes was shown. Furthermore, patients were characterised by the presence of low-grade inflammation, as demonstrated by increased infiltration of mucosal mast cells and eosinophils. A significant association between the expression level of several cell-to-cell adhesion proteins, the extent of increased permeability and the severity of low-grade inflammation was found. Conclusions: These findings challenge the classical paradigm that patients with FD show no structural changes in the gastrointestinal tract. We suggest that impaired intestinal barrier function is a pathophysiological mechanism in FD. Thus, restoration of intestinal barrier integrity may be a potential therapeutic target for treating patients with FD.
Article
Symptoms that can be attributed to the gastroduodenal region represent one of the main subgroups among functional gastrointestinal disorders. A slightly modified classification into four categories is proposed. B1) Functional dyspepsia (FD), characterized by one or more of: postprandial fullness, early satiation, epigastric pain, epigastric burning that are unexplained after a routine clinical evaluation, includes two subcategories: Postprandial Distress Syndrome (PDS) that is characterized by meal-induced dyspeptic symptoms and Epigastric Pain Syndrome (EPS) that does not occur exclusively postprandially. The two subgroups may overlap. B2) Belching disorders, defined as audible escapes of air from the esophagus or the stomach, is classified into 2 subcategories, depending on the origin of the refluxed gas as detected by intraluminal impedance measurement belching: gastric and supragastric belch. B3) Nausea and vomiting disorders include three subcategories: chronic nausea and vomiting syndrome (CNVS); cyclic vomiting syndrome (CVS); cannabinoid hyperemesis syndrome (CHS). B4) Rumination syndrome.
Article
Background Correct neuronal identification is essential to study neurons in health and disease. Although commonly used as pan-neuronal marker, HuC/D's expression pattern varies substantially between healthy and (patho)physiological conditions. This heterogenic labeling has received very little attention. We sought to investigate the subcellular HuC/D localization in enteric neurons in different conditions.Methods The localization of neuronal RNA-binding proteins HuC/D was investigated by immunohistochemistry in the mouse myenteric plexus using different toxins and caustic agents. Preparations were also stained with Sox10 and glial fibrillary acidic protein (GFAP) antibodies to assess enteric glial cell appearance.Key ResultsMechanically induced tissue damage, interference with the respiratory chain and oxygen (O2) deprivation increased nuclear HuC/D immunoreactivity. This effect was paralleled by a distortion of the GFAP-labeled glial network along with a loss of Sox10 expression and coincided with the activation of a non-apoptotic genetic program. Chemically induced damage and specific noxious stimuli did not induce a change in HuC/D immunoreactivity, supporting the specific nature of the nuclear HuC/D localization.Conclusions & InferencesHuC/D is not merely a pan-neuronal marker but its subcellular localization also reflects the condition of a neuron at the time of fixation. The functional meaning of this change in HuC/D localization is not entirely clear, but disturbance in O2 supply in combination with the support of enteric glial cells seems to play a crucial role. The molecular consequence of changes in HuC/D expression needs to be further investigated.
Article
Functional dyspepsia (FD), defined by unexplained pain or discomfort centred in the upper abdomen, is common. Diagnosis and treatment of FD based on the symptom-based Rome criteria remains challenging. Recently, eosinophilia in the duodenum has been implicated in the pathophysiology of FD in adults, specifically increased eosinophils in early satiety and postprandial distress but the association remains controversial. The aim of this study was to characterize upper gastrointestinal tract pathology, specifically duodenal eosinophilia, in an Australian cohort of patients with FD. Patients prospectively referred for an upper gastrointestinal endoscopy (n=55; mean age, 49.6 years; 61.8% female) were stratified to FD cases (n=33) and controls (n=22) using Rome II criteria. All subjects completed a validated Bowel Symptom Questionnaire. The eosinophil count per mm(2) in the duodenal bulb (D1) and second part (D2) was assessed and H. pylori status determined by gastric histology. Associations with clinical symptoms were assessed. Cases and controls were demographically similar. Duodenal eosinophilia was significantly increased in subjects experiencing early satiety (p=0.01) and post prandial fullness (p=0.001). This association was seen in D2 but not D1. Abdominal pain was associated with eosinophilia in both D1 (p=0.02) and D2 (p=0.005). Smoking was also associated with higher eosinophil counts in D2 (p=0.007) and symptoms of early satiety (p=0.02). Duodenal eosinophilia occurs in a subset of FD. The potential role of duodenal eosinophils in FD has implications for diagnosis and therapeutic trials.
Article
The aetiology of necrotising enterocolitis (NEC) is unknown, but luminal factors and epithelial leakiness appear critical triggers of an inflammatory cascade. A separate finding has been suggested in mouse models, in which disruption of glial cells in the myenteric plexus induced a severe NEC-like lesion. We have thus looked for evidence of neuroglial abnormality in NEC. We studied full-thickness resected specimens from 20 preterm infants with acute NEC and from 13 control infants undergoing resection for other indications. Immunohistochemical analysis was performed for immunological (CD3, syndecan-1, human leucocyte antigen-DR), neural (glial fibrillary acidic protein [GFAP], nerve growth factor receptor, neurofilament protein, neuron-specific enolase), and functional markers (Ki67), and for potential inflammatory regulators (interleukin-12, transforming growth factor-β, CCL20, CCR6). Expression of the chemokine CCL20 and its receptor CCR6 was significantly upregulated in myenteric plexus in NEC, with CCL20 strongly expressed by glial cells. In 9 of 20 cases with NEC, myenteric plexus architecture and GFAP+ glial cells were normal, with preserved submucosal and mucosal innervation; however, 11 cases showed disrupted myenteric plexus architecture, reduced GFAP expression, and loss of submucosal and mucosal innervation. Persistent abnormalities were identified in the 2 infants who had ongoing inflammation at ileostomy closure. Our findings identified heterogeneity among patients with NEC. Approximately half showed evidence of marked neural abnormality extending from the deeper layers of the intestine, associated with glial activation and myenteric plexus disruption. The factors that may activate enteric glia in this manner, potentially including bacterial products or viruses, remain to be determined.
Article
Background: Hyoscine butylbromide (HBB, Buscopan(®) ) is clinically used to treat intestinal cramps and visceral pain. Various studies, mainly on animal tissues, suggested that its antimuscarinic action is responsible for its spasmolytic effect. However, functional in vitro studies with human tissue have not been performed so far. Methods: We wanted to provide a comprehensive study on the mode of action of HBB in human intestinal samples and investigated HBB (1 nmol L(-1) -10 μmol L(-1)) effects on muscle activity with isometric force transducers and calcium imaging, on epithelial secretion with Ussing chamber technique and on enteric neurons using fast neuroimaging. Key results: Hyoscine butylbromide concentration dependently reduced muscle contractions, calcium mobilization, and epithelial secretion induced by the muscarinic agonist bethanechol with IC50 values of 429, 121, and 224 nmol L(-1), respectively. Forskolin-induced secretion was not altered by HBB. Cholinergic muscarinic muscle and epithelial responses evoked by electrical nerve stimulation were inhibited by 1-10 μmol L(-1) HBB. Moreover, HBB significantly reduced the bethanechol-induced action potential discharge in enteric neurons. Interestingly, we observed that high concentrations of HBB (10 μmol L(-1)) moderately decreased nicotinic receptor-mediated secretion, motility, and nerve activity. Conclusions & inferences: The results demonstrated the strong antimuscarinic action of HBB whereas the nicotinic antagonism at higher concentrations plays at most a moderate modulatory role. The muscle relaxing effect of HBB and its inhibition of muscarinic nerve activation likely explain its clinical use as an antispasmodic drug. Our results further highlight a so far unknown antisecretory action of HBB which warrants further clinical studies on its use in secretory disorders.