ArticlePDF AvailableLiterature Review

In vitro models for gluten toxicity: Relevance for celiac disease pathogenesis and development of novel treatment options

Authors:

Abstract and Figures

In genetically predisposed individuals, dietary gluten in wheat, rye and barley triggers celiac disease, a systemic autoimmune disorder hallmarked by an extensive small-bowel mucosal immune response. The current conception of celiac disease pathogenesis is that it involves components of both innate and adaptive immunity whose activation typically leads to small-bowel villous atrophy with crypt hyperplasia. Currently, the only effective treatment for celiac disease is a strict lifelong gluten-free diet excluding all wheat-, rye- and barley-containing food products. During the diet, the clinical symptoms improve and the small-bowel mucosal damage recovers, while re-introduction of gluten into the diet leads to re-appearance of the symptoms and deterioration of the small-bowel mucosal architecture. In view of the restricted nature of the diet, alternative treatment is warranted. Improved understanding of the molecular basis of celiac disease has enabled researchers to suggest other therapeutic approaches. Although there is no animal model reproducing all features of celiac disease, the use of in vitro approaches including a variety of cell lines and the celiac patient small-bowel mucosal biopsy organ culture has generated knowledge about pathogenesis of celiac disease. In these culture systems, gluten induces different effects that can be quantified, thus also enabling studies concerning the efficacy of candidate therapeutic compounds for celiac disease. This review describes the intestinal epithelial cell models, celiac patient T-cell lines and clones, as well as the small-bowel mucosal organ culture methods widely used in studies of celiac disease, and summarizes the major findings obtained with these systems.
Content may be subject to copyright.
Minireview
In vitro models for gluten toxicity: relevance for celiac disease
pathogenesis and development of novel treatment options
Katri Lindfors1, Tiina Rauhavirta1, Satumarja Stenman1, Markku Ma
¨ki1and Katri Kaukinen2,3
1
Pediatric Research Center, University of Tampere and Tampere University Hospital;
2
School of Medicine, University of Tampere, 33014
Tampere;
3
Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, 33521 Tampere, Finland
Corresponding author: Dr Katri Lindfors, Pediatric Research Center, School of Medicine, Finn-Medi 3, University of Tampere, Tampere
33014, Finland. Email: katri.lindfors@uta.fi
Abstract
In genetically predisposed individuals, dietary gluten in wheat, rye and barley triggers celiac disease, a systemic autoimmune
disorder hallmarked by an extensive small-bowel mucosal immune response. The current conception of celiac disease
pathogenesis is that it involves components of both innate and adaptive immunity whose activation typically leads to
small-bowel villous atrophy with crypt hyperplasia. Currently, the only effective treatment for celiac disease is a strict
lifelong gluten-free diet excluding all wheat-, rye- and barley-containing food products. During the diet, the clinical
symptoms improve and the small-bowel mucosal damage recovers, while re-introduction of gluten into the diet leads to
re-appearance of the symptoms and deterioration of the small-bowel mucosal architecture. In view of the restricted nature
of the diet, alternative treatment is warranted. Improved understanding of the molecular basis of celiac disease has
enabled researchers to suggest other therapeutic approaches. Although there is no animal model reproducing all features
of celiac disease, the use of in vitro approaches including a variety of cell lines and the celiac patient small-bowel mucosal
biopsy organ culture has generated knowledge about pathogenesis of celiac disease. In these culture systems, gluten
induces different effects that can be quantified, thus also enabling studies concerning the efficacy of candidate
therapeutic compounds for celiac disease. This review describes the intestinal epithelial cell models, celiac patient T-cell
lines and clones, as well as the small-bowel mucosal organ culture methods widely used in studies of celiac disease, and
summarizes the major findings obtained with these systems.
Keywords: celiac disease, gluten, epithelium, T-cells, organ culture
Experimental Biology and Medicine 2012; 237: 119 –125. DOI: 10.1258/ebm.2011.011294
Introduction
Dietary gluten in wheat, rye and barley is the etiological
agent in celiac disease. In a subgroup of individuals carry-
ing the human leukocyte antigen (HLA) DQ2 or DQ8 mol-
ecules, ingestion of gluten leads to a small-bowel mucosal
immune response and development of celiac disease. The
disorder often presents with small-bowel villous atrophy,
crypt hyperplasia and immune activation, including the
production of disease-specific antibodies against gluten-
derived gliadin peptides and an endogenous protein trans-
glutaminase 2 (TG2).
1
The current conception of celiac
disease pathogenesis is that it involves components of
both innate and adaptive immunity. Gluten contains a
high amount of repetitive glutamine- and proline-rich
sequences, making it highly resistant to proteolytic degra-
dation by gastrointestinal enzymes.
2,3
This results in the per-
sistence of relatively large gluten-derived gliadin peptides
which predispose to activation of the small-bowel mucosal
immune system. Some of the gliadin peptides (the so-called
toxic peptides, for example p31–43) are able to induce
harmful effects on the small-intestinal epithelium through
interleukin (IL)-15, whose overexpression eventually leads
to epithelial cell destruction.
4,5
Other gliadin peptides, the
immunodominant ones ( for example p57– 68 and the
33-mer) in turn activate T-cell-mediated adaptive immunity
in the mucosa. A key step in this process is the post-
translational modification of immunogenic gliadin peptides
by TG2. TG2 has the capacity to deamidate distinct gluta-
mine residues in gliadin peptides to glutamic acid, which
favors their interaction with the celiac-type HLA DQ2,
thereby promoting the activation of T-cells.
3,6,7
Similar to other autoimmune disorders, celiac disease is
self-perpetuating if the specific exogenous trigger, gluten,
is not removed. Its removal by a strict gluten-free diet
ISSN: 1535-3702
Copyright #2012 by the Society for Experimental Biology and Medicine
Experimental Biology and Medicine 2012; 237: 119 125
leads to recovery of the small-bowel mucosal lesion and dis-
appearance of the clinical symptoms. Re-introduction of
gluten into the diet, on the other hand, results in the
re-appearance of clinical symptoms and deterioration of
the small-bowel mucosal architecture. Today, the only effec-
tive treatment for celiac disease is a lifelong gluten-free diet
excluding all wheat-, rye- and barley-containing food pro-
ducts. As the diet is restricted and often burdensome,
novel treatment options would be highly welcomed by
celiac patients. During recent years, several lines of research
have in fact aimed to develop alternative therapies.
Approaches include blocking of innate and adaptive
immune components as well as the function of TG2.
8
Another prominent strategy is to hydrolyze gluten by
exogenous enzyme supplements designed for this
purpose. This promising strategy is based on the finding
that distinct peptidases or their combinations from different
sources are able to hydrolyze gliadin and gliadin peptides
in vitro.
3,9
Indeed, data are available to indicate that distinct
peptidase is capable of accelerating the degradation of
gluten in the gastrointestinal system, closely mimicking
in vivo digestion and suggesting that gluten toxicity might
be eliminated by such an approach.
10
Regardless of the progress made in understanding the
pathogenesis of celiac disease, the most conspicuous limit-
ation in the field is the lack of a functional disease-specific
animal model. Such a model would also be highly appreci-
ated among researchers developing novel treatment strat-
egies for the disease, as the initial testing of novel drug
candidates could then be conducted in animals. A number
of attempts have been made to create an animal model for
celiac disease by various approaches. These include the gen-
eration of transgenic mice expressing the celiac-type human
HLA DQ2 or DQ8
11 – 14
and introduction of celiac-type anti-
bodies in mice,
15 – 17
but none of the animals used have
developed a full-blown villous atrophy together with
crypt hyperplasia that are characteristic for untreated
celiac disease. Gluten-dependent small-intestinal mucosal
damage can be induced in mice
18
and has been described
in rhesus macaques,
19
but these two models are not depen-
dent on celiac-type HLA and their applicability as models
for celiac disease has thus to be considered with caution.
Very recently, DePaolo et al.
20
reported that gliadin-fed
humanized HLA-DQ8 mice that overexpress IL-15 in the
lamina propria (DQ8-D
d
-IL-15 transgenic mice) present
with a condition resembling early developing celiac
disease, where intestinal inflammation is present but the
mucosal architecture remains normal. This promising
model may prove useful in the future, although its applica-
bility needs to be verified in further studies.
Regardless of numerous attempts, the animal model for
celiac disease that reproduces all the aspects of the disorder
still awaits development. However, the use of in vitro
approaches including a variety of cell lines and the celiac
patient small-bowel mucosal biopsy organ culture has gen-
erated knowledge about pathogenesis of celiac disease and
enabled the initial testing of novel treatment options for
celiac disease. This review describes the cell-based and
organ culture methods widely used in celiac disease and
summarizes the major findings obtained with these systems.
Epithelial cell culture models
After ingestion and progress to the small intestine, the
gluten-derived gliadin peptides come into contact with the
small-bowel mucosal epithelium. In untreated celiac
disease, small-bowel mucosal epithelial cells proliferate
more rapidly without differentiating to enterocytes, and
their turnover by apoptosis is augmented. Moreover, the
epithelial junctional integrity is compromised
21,22
and the
gliadin peptides therefore gain access to the lamina
propria. However, it is not well understood how gluten-
derived gliadin peptides modulate the biology of the epi-
thelium and how they cross the epithelial barrier prior to
substantial mucosal remodeling during the early stages of
the disorder. For all of the above-mentioned reasons,
study of the intestinal epithelium in the context of celiac
disease is thus manifestly relevant. There are several intesti-
nal epithelial cell lines available, but two of those mostly
used in celiac disease research are described here.
T84 cells
T84 cells are derived from a lung metastasis of a colon
cancer, and the cell line was first established as a model
for studying intestinal epithelial chloride transport. When
grown on permeable supports, the cells form confluent
polarized layers with high transepithelial resistance, and
they evince a chloride secretory response to different
chemicals.
23
In addition, the polarized T84 cell layers are
characterized with well-delineated intracellular junctions
including circumferential tight junctions.
24
For the above-
mentioned reason, the cell line, although of colonic origin,
is widely used in studying intestinal epithelial permeability
to macromolecules and ions even though the cells do not
present with features of intestinal enterocyte-type differen-
tiation in such cultures. However, when T84 cells are cul-
tured three-dimensionally in collagen in the presence of
either fibroblasts or transforming growth factor
b
, they
differentiate to intestinal enterocytes as assessed by mor-
phological and biochemical criteria.
25
T84 cells have been applied in research into celiac disease
pathogenesis, for instance in studying the innate immune
reactions elicited by the toxic gliadin peptides. It has been
shown that the toxic gliadin peptide p31 43 creates cellular
stress by inducing the production of reactive oxygen
species
26
and Fas-dependent apoptosis.
27
Furthermore, it
has been shown that untreated celiac disease patients’
serum antibodies induce proliferation and reduce differen-
tiation of the three-dimensionally cultured T84 cells
28
and
increase the permeability of the T84 cell epithelial mono-
layer.
29
Moreover, Bethune et al.
30
have demonstrated that
interferon (IFN)-
g
derived from gluten-stimulated celiac
patientspecific intestinal T-cells enhances the passage of
gluten through the T84 epithelial layer.
Caco-2 cells
The Caco-2 cell line was originally obtained from a relatively
well-differentiated human colon adenocarcinoma. When
reaching confluence, the Caco-2 cells differentiate
.................................. ..... ..... ..... ..... ..... ...... .............................. ..... ...... ..... ..... ..... ..... ..... ..... ...... ..
120 Experimental Biology and Medicine Volume 237 February 2012
spontaneously.
31
In such cultures, the cells are polarized
and possess an ultrastructural morphology of differentiated
enterocytes with an apical surface covered by microvilli.
32
In
the monolayer, the cells are joined together through apical
junctional complexes which include tight junctions and des-
mosomes.
33
Postconfluent cultures are hallmarked by the
emergence of domes in which cells express sucrase isomal-
tase, alkaline phosphatase and aminopeptidase N,
enzymes typical for differentiated enterocytes.
33
Owing to
the above-mentioned characteristics, the Caco-2 cell line is
widely used as a model for the intestinal epithelial barrier.
Caco-2 cells probably comprise the most extensively used
epithelial cell line in studies related to celiac disease.
Gliadin or distinct gliadin peptides are reported to induce
actin rearrangement,
34
proliferation,
34,35
intracellular oxi-
dative imbalance
36
and apoptosis in Caco-2 cells.
37,38
Gliadin also inhibits the spontaneous differentiation of
these cells.
39
Such findings suggest that the celiac
disease-inducing agent gliadin itself could at least partly
account for the behavioral abnormalities of the small
bowel epithelial cells in celiac disease.
In view of the importance of the compromised small-
bowel epithelial barrier function in celiac disease pathogen-
esis, researchers have used the Caco-2 cells in studies
seeking to establish which factors could account for this.
At least gliadin and two proinflammatory cytokines abun-
dantly expressed in celiac disease, IFN-
g
and tumor necrosis
factor
a
, have been reported to affect the expression of
several epithelial junctional proteins (for example occludin
and ZO-1) and permeability in Caco-2 cells.
40 – 48
Caco-2 cells have also been used in studies clarifying how
individual gliadin peptides are processed in intestinal epi-
thelial cells. Using Caco-2 cells it has been found that both
the toxic p31 43 and the immunodominant p5768
gliadin peptides are taken up by the cells and interact
with the endocytosis compartment.
49
However, the toxic
and the immunodominant gliadin peptides seem to be dif-
ferentially processed in Caco-2 cells,
49
but at least the immu-
nodominant 33-mer gliadin peptide eventually crosses the
epithelial barrier to the luminal side.
50
These findings in
Caco-2 cells, together with others made in celiac patient
biopsies or mucosal organ cultures, have considerably
increased our understanding of gliadin processing in intes-
tinal epithelial cells, this presumably being crucial for our
understanding of celiac disease pathogenesis.
The epithelial modulating properties of yet another factor
possibly participating in celiac disease pathogenesis,
namely celiac disease antibodies, have also been studied
in Caco-2 cells. It has been shown that celiac patient anti-
bodies induce actin remodeling,
51
interfere with the
uptake of p3143
52
and modify its intracellular transport,
53
and further induce proliferation in Caco-2 cells.
51,54
Moreover, celiac patient IgA has been reported to increase
the epithelial permeability of both toxic ( p31 43) and
immunodominant ( p5768) gliadin peptides.
55
The Caco-2 cell line has also been used fairly extensively
in initial testing of novel treatment options for celiac
disease. The cell line has been used in studying the
potency of at least a tight junction modulator, zonulin
antagonist, Bifidobacteria, germinating cereal enzymes
and polymeric binders of gliadin as putative future treat-
ment options.
41 – 43,45,56 – 59
All the studies in question
demonstrated protective effects of the tested compounds
against gluten-induced effects in Caco-2 cells, but their effi-
cacy obviously need to be tested in other systems as well as
in patients. Taken together, the use of Caco-2 cells in celiac
disease research has shed significant light on the disease
pathogenesis and the proof-of-principle efficacy of novel
treatment options.
Celiac patient-derived T-cell lines and clones
The extensive evidence that CD4þT-cells, associated with
recognition of HLA DQ2 and DQ8 molecules, are markedly
involved in the pathogenesis of celiac disease has inspired
researchers to develop in vitro T-cell models. The celiac
disease-specific T-cells, raised against gluten, have been
obtained both from the small-intestinal mucosa
60 – 62
and
the peripheral blood
63 – 65
of patients suffering from celiac
disease. The HLA-DQ2 or DQ8 restricted T-cells isolated
from small-bowel mucosal biopsies are typically cultured
in vitro in the presence of stimulating gluten peptides
(T-cell lines), whereafter single, gluten-reactive monoclonal
cells (T-cell clones) can be further isolated and maintained
in the culture conditions. In contrast, peripheral blood
mononuclear cells are isolated from treated celiac disease
patient’s serum after a short-term in vivo gluten challenge.
66
In both cases, activation of T-cells is typically presented as
an increase in cell proliferation and IFN-
g
-predominant
cytokine production.
62,64,67
Most of the gut-derived T-cells do not recognize native
gluten peptides unless they are deamidated by tissue trans-
glutaminase,
6,7
a phenomenon which is thought to occur
also in in vivo conditions. The majority of studies conducted
with celiac patient T-cells have concentrated on identifi-
cation of gliadin epitopes such as diverse epitopes found
in immunodominant
a
-gliadin 33-mer;
3,68
however, intesti-
nal T-cell lines and clones have also been shown to recog-
nize deamidated rye secalin and barley hordein
peptides.
69,70
T-cell lines and clones have been especially
useful in the systematic characterization of T-cell reactive
gluten epitopes in wheat, rye and barley.
71 – 73
However,
patients seem to respond to distinct gluten peptides,
74,75
resulting in limitations when investigating the overall tox-
icity of gluten peptides. Application of several simultaneous
T-cell lines from different patients is thus probably needed
to overcome these challenges.
Similarly to intestinal epithelial cells, celiac patient T-cell
lines and clones have been used in testing newly developed
treatment forms. For instance, testing of different types of
modified gliadin or gluten peptides
76 – 79
and enzyme
therapy
3,42,80 – 83
as a potential novel treatment form has
been carried out in celiac patient T-cell lines or clones.
Small-bowel mucosal biopsy organ culture
The culturing of small-intestinal mucosal biopsies derived
ex vivo from celiac disease patients was originally intro-
duced by Browning and Trier.
84
The biopsies can be kept
.................................. ..... ..... ..... ..... ..... ...... .............................. ..... ...... ..... ..... ..... ..... ..... ..... ...... ..
Lindfors et al. In vitro models for gluten toxicity 121
viable in the organ culture system for 24 48 h
85
or even
longer, although there is no guarantee of the quality of
mucosal morphology after a long-term culture.
86
In prelimi-
nary studies, the organ culture method was used in an
opposite manner to the current practice. Culture conditions
were thought to mimic a gluten-free diet, so that the small-
bowel mucosal morphology improved after gluten was
excluded.
85 – 88
In contrast, the present organ culture
system is generated to illustrate the harmful effects of
gluten, gliadin or distinct gliadin peptides added to the
culture supernatant (Figure 1). Since the ex vivo biopsy
culture contains a wide variety of cell types expressed in
the intestinal mucosa in vivo, the method allows researchers
to investigate an extensive range of markers for gluten tox-
icity, all the way from the epithelium to activation of
immune components in the mucosal lamina propria. One of
the versatile benefits of the method lies in the fact that
various features characteristic for the disorder can be repro-
duced in biopsies from treated celiac disease patients, thus
allowing researchers to find mechanisms related to the
development of celiac disease from a healthy mucosa into
the active phase of the disease. In addition, some researchers
have suggested the organ culture method even as a poten-
tial tool for celiac disease diagnosis.
89,90
As the small-bowel mucosal biopsy organ culture system
has been used extensively in studies aiming to clarify the
pathogenesis of celiac disease, it would be impossible to
summarize the entire literature in this review. However,
since biopsies contain a number of different cell types, the
issues open to investigation in the system are numerous.
For instance, celiac disease researchers have studied the
immune mechanisms
4
as well as the contribution of different
cytokines
91
in this context. As an example, one of the major
findings related to celiac disease pathogenesis, namely the
importance of IL-15 during the disease process, was obtained
from studies conducted with the organ culture.
5,92 – 96
Considering that the small-bowel organ culture method
has been widely used in studies aiming to clarify the patho-
genesis of celiac disease, it is surprising that the method has
only been used in a few studies related to novel treatment
forms. To our knowledge, it has only been applied in study-
ing the therapeutic potential of a particular zonulin
antagonist
45
and gliadin-degrading proteolytic
enzymes,
42,81,97
with promising results.
Conclusions
In the absence of an animal model for celiac disease, research-
ers have used different cell lines and the celiac patient small-
bowel mucosal organ culture method in studying different
aspects of celiac disease pathogenesis and proof-of-concept
testing of novel treatment options which could in the future
replace the only effective treatment for celiac disease, the
gluten-free diet. Although widely used, all of the models pre-
sented in this review have their limitations and thus do not
always correlate with the circumstances in the human small
intestine in vivo. For example, the epithelial cell lines are not
derived from celiac disease patients but instead are of cancer-
ous origin. Moreover, the epithelial cells and celiac patient
T-cell lines and clones lack connection to other cell types
present in the intestine. In addition, the small-bowel organ
culture system lacks circulation, nervous system and connec-
tion to lymphatic organs. Furthermore, the organ culture
system is not a high-throughput method. In the future it
would thus be worthwhile to develop novel and improved
culture systems and to promote celiac disease-specific
models. However, even regardless of the above-mentioned
shortcomings, researchers have gained important infor-
mation and been able to addpieces to the celiac disease patho-
genesis puzzle using the models described in this review and
also other available model systems. Moreover, the studies
clarifying the efficacy of novel treatment options carried out
with the models described here have yielded encouraging
results and some potentially therapeutic compounds have
already entered phase II clinical trials.
Author contributions: KL, TR, SS, MM and KK all partici-
pated in the writing of the manuscript, and TR made the
figure.
ACKNOWLEDGEMENTS
The Celiac Disease Study Group have been financially sup-
ported by the Academy of Finland, the Sigrid Juselius
Foundation, the Foundation for Pediatric Research, the
Pirkanmaa Cultural Foundation, the Competitive Research
Funding of Tampere University Hospital, the Research
Fund of the Finnish Celiac Society and the European
Commission IAPP grant TRANSCOM (contract number
PIA-GA-2010-251506).
REFERENCES
1 Lindfors K, Ma
¨ki M, Kaukinen K. Transglutaminase 2-targeted
autoantibodies in celiac disease: pathogenetic players in addition to
diagnostic tools? Autoimmun Rev 2010;9:744– 9
2 Hausch F, Shan L, Santiago NA, Gray GM, Khosla C. Intestinal digestive
resistance of immunodominant gliadin peptides. Am J Physiol Gastrointest
Liver Physiol 2002;283:G996– G1003
3 Shan L, Molberg Ø, Parrot I, Hausch F, Filiz F, Gray GM, Sollid LM,
Khosla C. Structural basis for gluten intolerance in celiac sprue. Science
2002;297:22759
Figure 1 The principle of the small-bowel mucosal organ culture system
.................................. ..... ..... ..... ..... ..... ...... .............................. ..... ...... ..... ..... ..... ..... ..... ..... ...... ..
122 Experimental Biology and Medicine Volume 237 February 2012
4 Maiuri L, Ciacci C, Ricciardelli I, Vacca L, Raia V, Auricchio S, Picard J,
Osman M, Quaratino S, Londei M. Association between innate response
to gliadin and activation of pathogenic T cells in coeliac disease. Lancet
2003;362:30–7
5Hu
¨e S, Mention JJ, Monteiro RC, Zhang S, Cellier C, Schmitz J, Verkarre
V, Fodil N, Bahram S, Cerf-Bensussan N, Caillat-Zucman S. A direct role
for NKG2D/MICA interaction in villous atrophy during celiac disease.
Immunity 2004;21:367–7
6 Sjo
¨stro
¨m H, Lundin KE, Molberg O, Ko
¨rner R, McAdam SN, Anthonsen
D, Quarsten H, Nore
´n O, Roepstorff P, Thorsby E, Sollid LM.
Identification of a gliadin T-cell epitope in coeliac disease: general
importance of gliadin deamidation for intestinal T-cell recognition. Scand
J Immunol 1998;48:1115
7 Molberg O, McAdam SN, Korner R, Quarsten H, Kristiansen C, Madsen
L, Fugger L, Scott H, Nore
´n O, Roepstorff P, Lundin KE, Sjo
¨stro
¨mH,
Sollid LM. Tissue transglutaminase selectively modifies gliadin peptides
that are recognized by gut-derived T cells in celiac disease. Nat Med
1998;4:713–7
8 Sollid LM, Khosla C. Novel therapies for coeliac disease. J Intern Med
2011;269:60413
9 Gass J, Ehren J, Strohmeier G, Isaacs I, Khosla C. Fermentation,
purification, formulation, and pharmacological evaluation of a prolyl
endopeptidase from Myxococcus xanthus: implications for Celiac Sprue
therapy. Biotechnol Bioeng 2005;92:674– 84
10 Mitea C, Havenaar R, Drijfhout JW, Edens L, Dekking L, Koning F.
Efficient degradation of gluten by a prolyl endoprotease in a
gastrointestinal model: implications for coeliac disease. Gut
2008;57:25–32
11 D’Arienzo R, Stefanile R, Maurano F, Luongo D, Bergamo P, Mazzarella
G, Troncone R, Auricchio S, David C, Rossi M. A deregulated immune
response to gliadin causes a decreased villus height in DQ8 transgenic
mice. Eur J Immunol 2009;39:355261
12 de Kauwe AL, Chen Z, Anderson RP, Keech CL, Price JD, Wijburg O,
Jackson DC, Ladhams J, Allison J, McCluskey J. Resistance to
celiac disease in humanized HLA-DR3-DQ2-transgenic mice
expressing specific anti-gliadin CD4þT cells. J Immunol 2009;182:
7440–50
13 Senger S, Maurano F, Mazzeo MF, Gaita M, Fierro O, David CS,
Troncone R, Auricchio S, Siciliano RA, Rossi M. Identification of
immunodominant epitopes of alpha-gliadin in HLA-DQ8
transgenic mice following oral immunization. J Immunol
2005;175:808795
14 Du Pre
´MF, Kozijn AE, van Berkel LA, Ter Borg MN,
Lindenbergh-Kortleve D, Jensen LT, Kooy-Winkelaar Y, Koning F, Boon
L, Nieuwenhuis EE, Sollid LM, Fugger L, Samsom JN. Tolerance to
ingested deamidated gliadin in mice is maintained by splenic, type 1
regulatory T cells. Gastroenterology 2011;141:610–20
15 Smart CJ, Trejdosiewicz LK, Howdle PD. Specific circulating anti-gliadin
IgG-class antibody does not mediate intestinal enteropathy in gliadin-fed
mice. Int Arch Allergy Immunol 1992;97:160–6
16 Freitag T, Schulze-Koops H, Niedobitek G, Melino G, Schuppan D. The
role of the immune response against tissue transglutaminase in the
pathogenesis of coeliac disease. Autoimmun Rev 2004;3:12– 20
17 Di Niro R, Sblattero D, Florian F, Stebel M, Zentilin L, Giacca M,
Villanacci V, Galletti A, Not T, Ventura A, Marzari R. Anti-idiotypic
response in mice expressing human autoantibodies. Mol Immunol
2008;45:178291
18 Freitag TL, Rietdijk S, Junker Y, Popov Y, Bhan AK, Kelly CP, Terhorst C,
Schuppan D. Gliadin-primed CD4þCD45RBlowCD252T cells drive
gluten-dependent small intestinal damage after adoptive transfer into
lymphopenic mice. Gut 2009;58:1597605
19 Bethune MT, Borda JT, Ribka E, Liu MX, Phillippi-Falkenstein K,
Jandacek RJ, Doxiadis GG, Gray GM, Khosla C, Sestak K. A non-human
primate model for gluten sensitivity. PLoS One 2008;3:e1614
20 DePaolo RW, Abadie V, Tang F, Fehlner-Peach H, Hall JA, Wang W,
Marietta EV, Kasarda DD, Waldmann TA, Murray JA, Semrad C, Kupfer
SS, Belkaid Y, Guandalini S, Jabri B. Co-adjuvant effects of retinoic acid
and IL-15 induce inflammatory immunity to dietary antigens. Nature
2011;471:2204
21 Schulzke JD, Schulzke I, Fromm M, Riecken EO. Epithelial barrier and
ion transport in coeliac sprue: electrical measurements on intestinal
aspiration biopsy specimens. Gut 1995;37:777– 82
22 Schulzke JD, Bentzel CJ, Schulzke I, Riecken EO, Fromm M. Epithelial
tight junction structure in the jejunum of children with acute and treated
celiac sprue. Pediatr Res 1998;43:435– 41
23 Dharmsathaphorn K, McRoberts JA, Mandel KG, Tisdale LD, Masui H.
A human colonic tumor cell line that maintains vectorial electrolyte
transport. Am J Physiol Gastrointest Liver Physiol 1984;246:G204– 8
24 Madara JL, Stafford J, Dharmsathaphorn K, Carlson S. Structural
analysis of a human intestinal epithelial cell line. Gastroenterology
1987;92:113345
25 Halttunen T, Marttinen A, Rantala I, Kainulainen H, Ma
¨ki M. Fibroblasts
and transforming growth factor beta induce organization and
differentiation of T84 human epithelial cells. Gastroenterology
1996;111:125262
26 Luciani A, Villella VR, Vasaturo A, Giardino I, Pettoello-Mantovani M,
Guido S, Cexus ON, Peake N, Londei M, Quaratino S, Maiuri L.
Lysosomal accumulation of gliadin p31– 43 peptide induces oxidative
stress and tissue transglutaminase-mediated PPARgamma
downregulation in intestinal epithelial cells and coeliac mucosa. Gut
2010;59:311–9
27 Maiuri L, Ciacci C, Ricciardelli I, Vacca L, Raia V, Rispo A, Griffin M,
Issekutz T, Quaratino S, Londei M. Unexpected role of surface
transglutaminase type II in celiac disease. Gastroenterology
2005;129:140013
28 Halttunen T, Ma
¨ki M. Serum immunoglobulin A from patients with
celiac disease inhibits human T84 intestinal crypt epithelial cell
differentiation. Gastroenterology 1999;116:566–72
29 Zanoni G, Navone R, Lunardi C, Tridente G, Bason C, Sivori S, Beri R,
Dolcino M, Valletta E, Corrocher R, Puccetti A. In celiac disease, a subset
of autoantibodies against transglutaminase binds toll-like receptor 4 and
induces activation of monocytes. PLoS Med 2006;3:e358
30 Bethune MT, Siegel M, Howles-Banerji S, Khosla C. Interferon-gamma
released by gluten-stimulated celiac disease-specific intestinal T cells
enhances the transepithelial flux of gluten peptides. Pharmacol Exp Ther
2009;329:65768
31 Zweibaum A, Triadou N, Kedinger M, Augeron C, Robine-Le
´on S, Pinto
M, Rousset M, Haffen K. Sucrase-isomaltase: a marker of foetal and
malignant epithelial cells of the human colon. Int J Cancer
1983;32:407–12
32 Bolte G, Wolburg H, Beuermann K, Stocker S, Stern M. Specific
interaction of food proteins with apical membranes of the human
intestinal cell lines Caco-2 and T84. Clin Chim Acta 1998;270:15167
33 Pinto M, Robine-Leon S, Appay M-D, Kedinger M, Traidou N, Dussaulx
E, Lacroix B, Simon Assmann P, Haffen K, Fog J, Zweibaum A.
Enterocyte-like differentiation and polarization of the human colon
carcinoma cell line CaCo-2 in culture. Biol Cell 1983;47:32330
34 Barone MV, Gimigliano A, Castoria G, Paolella G, Maurano F, Paparo F,
Maglio M, Mineo A, Miele E, Nanayakkara M, Troncone R, Auricchio S.
Growth factor-like activity of gliadin, an alimentary protein: implications
for coeliac disease. Gut 2007;56:480 8
35 Barone MV, Zanzi D, Maglio M, Nanayakkara M, Santagata S, Lania G,
Miele E, Ribecco MT, Maurano F, Auricchio R, Gianfrani C, Ferrini S,
Troncone R, Auricchio S. Gliadin-mediated proliferation and innate
immune activation in celiac disease are due to alterations in vesicular
trafficking. PLoS One 2011;6:e17039
36 Rivabene R, Mancini E, De Vincenzi M. In vitro cytotoxic effect of wheat
gliadin-derived peptides on the Caco-2 intestinal cell line is associated
with intracellular oxidative imbalance: implications for coeliac disease.
Biochim Biophys Acta 1999;1453:152–60
37 Giovannini C, Sanchez M, Straface E, Scazzocchio B, Silano M, De
Vincenzi M. Induction of apoptosis in caco-2 cells by wheat gliadin
peptides. Toxicology 2000;145:63– 71
38 Sakly W, Thomas V, Quash G, El Alaoui S. A role for tissue
transglutaminase in alpha-gliadin peptide cytotoxicity. Clin Exp Immunol
2006;146:5508
39 Giovannini C, Maiuri L, De Vincenzi M. Cytotoxic effect of
prolamin-derived peptides on in vitro cultures of cell line Caco-2:
implications for coeliac disease. Toxicol In Vitro 1995;9:251–5
40 Sander GR, Cummins AG, Henshall T, Powell BC. Rapid disruption of
intestinal barrier function by gliadin involves altered expression of apical
junctional proteins. FEBS Lett 2005;579:4851– 5
41 Lindfors K, Blomqvist T, Juuti-Uusitalo K, Stenman S, Vena
¨la
¨inen J, Ma
¨ki
M, Kaukinen K. Live probiotic Bifidobacterium lactis bacteria inhibit the
.................................. ..... ..... ..... ..... ..... ...... .............................. ..... ...... ..... ..... ..... ..... ..... ..... ...... ..
Lindfors et al. In vitro models for gluten toxicity 123
toxic effects induced by wheat gliadin in epithelial cell culture. Clin Exp
Immunol 2008;152:5528
42 Stenman S, Vena
¨la
¨inen JI, Lindfors K, Auriola S, Mauriala T,
Kaukovirta-Norja A, Jantunen A, Laurila K, Qiao SW, Sollid LM,
Ma
¨nnisto PT, Kaukinen K, Ma
¨ki M. Enzymatic detoxification of gluten
by germinating wheat proteases: implications for new treatment of celiac
disease. Ann Med 2009;41:390400
43 Stenman S, Lindfors K, Vena
¨la
¨inen JI, Hautala A, Ma
¨nnisto
¨PT,
Garcia-Horsman JA, Kaukovirta-Norja A, Auriola S, Mauriala T, Ma
¨ki
M, Kaukinen K. Degradation of coeliac disease-inducing rye secalin by
germinating cereal enzymes: diminishing toxic effects in intestinal
epithelial cells. Clin Exp Immunol 2010;161:2429
44 Juuti-Uusitalo K, Lindfors K, Ma
¨ki M, Patrikainen M, Isola J, Kaukinen
K. Inhibition of epithelial growth factor receptor signalling does not
preserve epithelial barrier function after in vitro gliadin insult. Scand J
Gastroenterol 2009;44:820– 5
45 Drago S, El Asmar R, Di Pierro M, Grazia Clemente M, Tripathi A,
Sapone A, Thakar M, Iacono G, Carroccio A, D’Agate C, Not T, Zampini
L, Catassi C, Fasano A. Gliadin, zonulin and gut permeability: effects on
celiac and non-celiac intestinal mucosa and intestinal cell lines. Scand J
Gastroenterol 2006;41:408– 19
46 Perry I, Tselepis C, Hoyland J, Iqbal TH, Scott D, Sanders SA, Cooper BT,
Jankowski JA. Reduced cadherin/catenin complex expression in celiac
disease can be reproduced in vitro by cytokine stimulation. Lab Invest
1999;79:148999
47 Ciccocioppo R, Finamore A, Ara C, Di Sabatino A, Mengheri E,
Corazza GR. Altered expression, localization, and phosphorylation of
epithelial junctional proteins in celiac disease. Am J Clin Pathol
2006;125:50211
48 Tripathi A, Lammers KM, Goldblum S, Shea-Donohue T, Netzel-Arnett
S, Buzza MS, Antalis TM, Vogel SN, Zhao A, Yang S, Arrietta MC,
Meddings JB, Fasano A. Identification of human zonulin, a physiological
modulator of tight junctions, as prehaptoglobin-2. Proc Natl Acad Sci
2009;106:16799804
49 Barone MV, Nanayakkara M, Paolella G, Maglio M, Vitale V, Troiano R,
Ribecco MT, Lania G, Zanzi D, Santagata S, Auricchio R, Troncone R,
Auricchio S. Gliadin peptide P31 43 localises to endocytic vesicles and
interferes with their maturation. PLoS One 2010;5:e12246
50 Schumann M, Richter JF, Wedell I, Moos V, Zimmermann-Kormann M,
Schneider T, Daum S, Zeitz M, Fromm M, Schulzke JD. Mechanisms of
epithelial translocation of the alpha2-gliadin-33mer in coeliac sprue. Gut
2008;57:747–54
51 Barone MV, Caputo I, Ribecco MT, Maglio M, Marzari R, Sblattero D,
Troncone R, Auricchio S, Esposito C. Humoral immune response to
tissue transglutaminase is related to epithelial cell proliferation in celiac
disease. Gastroenterology 2007;132:1245– 53
52 Caputo I, Barone MV, Lepretti M, Martucciello S, Nista I, Troncone R,
Auricchio S, Sblattero D, Esposito C. Celiac anti-tissue transglutaminase
antibodies interfere with the uptake of alpha gliadin peptide 31 43 but
not of peptide 57– 68 by epithelial cells. Biochim Biophys Acta
2010;1802:71727
53 Lu
¨bbing N, Barone MV, Rudloff S, Troncone R, Auricchio S, Zimmer KP.
Correction of gliadin transport within enterocytes through celiac disease
serum. Pediatr Res 2011;70:357– 62
54 Ferrara F, Quaglia S, Caputo I, Esposito C, Lepretti M, Pastore S, Giorgi
R, Martelossi S, Dal Molin G, Di Toro N, Ventura A, Not T.
Anti-transglutaminase antibodies in non-coeliac children suffering from
infectious diseases. Clin Exp Immunol 2010;159:217 23
55 Rauhavirta T, Qiao SW, Jiang Z, Myrsky E, Loponen J, Korponay-Szabo
´
IR, Salovaara H, Garcia-Horsman JA, Vena
¨la
¨inen J, Ma
¨nnisto
¨PT,
Collighan R, Mongeot A, Griffin M, Ma
¨ki M, Kaukinen K, Lindfors K.
Epithelial transport and deamidation of gliadin peptides: a role for
coeliac disease patient immunoglobulin A. Clin Exp Immunol
2011;164:12736
56 Laparra JM, Sanz Y. Bifidobacteria inhibit the inflammatory response
induced by gliadins in intestinal epithelial cells via modifications
of toxic peptide generation during digestion. J Cell Biochem
2010;109:8017
57 De Palma G, Cinova J, Stepankova R, Tuckova L, Sanz Y. Pivotal
Advance: Bifidobacteria and Gram-negative bacteria differentially
influence immune responses in the proinflammatory milieu of celiac
disease. J Leuk Biol 2010;87:76578
58 Olivares M, Laparra M, Sanz Y. Influence of Bifidobacterium longum CECT
7347 and gliadin peptides on intestinal epithelial cell proteome. J Agric
Food Chem 2011;59:7666– 71
59 Pinier M, Verdu EF, Nasser-Eddine M, David CS, Ve
´zina A, Rivard N,
Leroux JC. Polymeric binders suppress gliadin-induced toxicity in the
intestinal epithelium. Gastroenterology 2009;136:288– 98
60 van de Wal Y, Kooy YM, van Veelen PA, Pena SA, Mearin LM, Molberg
O, Lundin KE, Sollid LM, Mutis T, Benckhuijsen WE, Drijfhout JW,
Koning F. Small intestinal T cells of celiac disease patients recognize a
natural pepsin fragment of gliadin. Proc Natl Acad Sci USA
1998;95:100504
61 Lundin KE, Scott H, Hansen T, Paulsen G, Halstensen TS, Fausa O,
Thorsby E, Sollid LM. Gliadin-specific, HLA-DQ(alpha 10501, beta
10201) restricted T cells isolated from the small intestinal mucosa of
celiac disease patients. J Exp Med 1993;178:187– 96
62 Ellis HJ, Pollock EL, Engel W, Fraser JS, Rosen-Bronson S, Wieser H,
Ciclitira PJ. Investigation of the putative immunodominant T cell
epitopes in coeliac disease. Gut 2003;52:212– 7
63 Ra
´ki M, Fallang LE, Brottveit M, Bergseng E, Quarsten H, Lundin KE,
Sollid LM. Tetramer visualization of gut-homing gluten-specific T cells in
the peripheral blood of celiac disease patients. Proc Natl Acad Sci
2007;104:28316
64 Anderson RP, van Heel DA, Tye-Din JA, Barnardo M, Salio M, Jewell DP,
Hill AV. T cells in peripheral blood after gluten challenge in coeliac
disease. Gut 2005;54:121723
65 Gjertsen HA, Lundin KE, Sollid LM, Eriksen JA, Thorsby E. T cells
recognize a peptide derived from alpha-gliadin presented by the celiac
disease-associated HLA-DQ (alpha 10501, beta 10201) heterodimer.
Hum Immunol 1994;39:24352
66 Anderson RP, Degano P, Godkin AJ, Jewell DP, Hill AV. In vivo antigen
challenge in celiac disease identifies a single transglutaminase-modified
peptide as the dominant A-gliadin T-cell epitope. Nat Med 2000;6:337– 42
67 Nilsen EM, Gjertsen HA, Jensen K, Brandtzaeg P, Lundin KE. Gluten
activation of peripheral blood T cells induces a Th0-like cytokine pattern
in both coeliac patients and controls. Clin Exp Immunol 1996;103:295–303
68 Qiao SW, Bergseng E, Molberg Ø, Xia J, Fleckenstein B, Khosla C, Sollid
LM. Antigen presentation to celiac lesion-derived T cells of a 33-mer
gliadin peptide naturally formed by gastrointestinal digestion. J Immunol
2004;173:175762
69 Vader LW, Stepniak DT, Bunnik EM, Kooy YM, de Haan W, Drijfhout
JW, Van Veelen PA, Koning F. Characterization of cereal toxicity for
celiac disease patients based on protein homology in grains.
Gastroenterology 2003;125:1105– 13
70 Kilmartin C, Wieser H, Abuzakouk M, Kelly CP, Jackson J, Feighery C.
Intestinal T cell responses to cereal proteins in celiac disease. Dig Dis Sci
2006;51:20029
71 Beissbarth T, Tye-Din JA, Smyth GK, Speed TP, Anderson RP. A
systematic approach for comprehensive T-cell epitope discovery using
peptide libraries. Bioinformatics 2005;21:i29-i37
72 Arentz-Hansen H, McAdam SN, Molberg Ø, Fleckenstein B, Lundin KE,
Jørgensen TJ, Jung G, Roepstorff P, Sollid LM. Celiac lesion T cells
recognize epitopes that cluster in regions of gliadins rich in proline
residues. Gastroenterology 2002;123:803– 9
73 Shan L, Qiao SW, Arentz-Hansen H, Molberg Ø, Gray GM, Sollid LM,
Khosla C. Identification and analysis of multivalent proteolytically
resistant peptides from gluten: implications for celiac sprue. J Proteome
Res 2005;4:1732 41
74 Vader W, Kooy Y, Van Veelen P, De Ru A, Harris D, Benckhuijsen W,
Pen˜a S, Mearin L, Drijfhout JW, Koning F. The gluten response in
children with celiac disease is directed toward multiple gliadin and
glutenin peptides. Gastroenterology 2002;122:1729– 37
75 Lundin KE, Sollid LM, Anthonsen D, Noren O, Molberg O, Thorsby E,
Sjo
¨stro
¨m H. Heterogeneous reactivity patterns of HLA-DQ-restricted,
small intestinal T-cell clones from patients with celiac disease.
Gastroenterology 1997;112:752– 9
76 Gianfrani C, Siciliano RA, Facchiano AM, Camarca A, Mazzeo MF,
Costantini S, Salvati VM, Maurano F, Mazzarella G, Iaquinto G, Bergamo
P, Rossi M. Transamidation of wheat flour inhibits the response to
gliadin of intestinal T cells in celiac disease. Gastroenterology
2007;133:7809
77 Huan J, Meza-Romero R, Mooney JL, Vandenbark AA, Offner H,
Burrows GG. Single-chain recombinant HLA-DQ2.5/peptide molecules
.................................. ..... ..... ..... ..... ..... ...... .............................. ..... ...... ..... ..... ..... ..... ..... ..... ...... ..
124 Experimental Biology and Medicine Volume 237 February 2012
block
a
2-gliadin-specific pathogenic CD4þT-cell proliferation and
attenuate production of inflammatory cytokines: a potential therapy for
celiac disease. Mucosal Immunol 2011;4:112– 20
78 Kapoerchan VV, Wiesner M, Overhand M, van der Marel GA, Koning F,
Overkleeft HS. Design of azidoproline containing gluten peptides to
suppress CD4þT-cell responses associated with celiac disease. Bioorg
Med Chem 2008;16:205362
79 Xia J, Bergseng E, Fleckenstein B, Siegel M, Kim CY, Khosla C, Sollid LM.
Cyclic and dimeric gluten peptide analogues inhibiting DQ2-mediated
antigen presentation in celiac disease. Bioorg Med Chem 2007;15:6565– 73
80 Gass J, Bethune MT, Siegel M, Spencer A, Khosla C. Combination
enzyme therapy for gastric digestion of dietary gluten in patients with
celiac sprue. Gastroenterology 2007;133:472– 80
81 Marti T, Molberg O, Li Q, Gray GM, Khosla C, Sollid LM. Prolyl
endopeptidase-mediated destruction of T cell epitopes in whole gluten:
chemical and immunological characterization. J Pharmacol Exp Ther
2005;312:19–26
82 Stepniak D, Spaenij-Dekking L, Mitea C, Moester M, de Ru A,
Baak-Pablo R, van Veelen P, Edens L, Koning F. Highly efficient gluten
degradation with a newly identified prolyl endoprotease: implications
for celiac disease. Am J Physiol Gastrointest Liver Physiol
2006;291:G621–G29
83 Siegel M, Bethune MT, Gass J, Ehren J, Xia J, Johannsen A, Stuge TB,
Gray GM, Lee PP, Khosla C. Rational design of combination enzyme
therapy for celiac sprue. Chem Biol 2006;13:649 58
84 Browning TH, Trier JS. Organ culture of mucosal biopsies of human
small intestine. J Clin Invest 1969;48:1423 32
85 Jos J, Lenoir G, Ritis GD, Rey J. In vitro pathogenetic studies of coeliac
disease. Effects of protein digests on coeliac intestinal biopsy specimens
maintained in culture for 48 hours. Scand J Gastroenterol 1975;10:1218
86 Falchuk ZM, Gebhard RL, Sessoms C, Strober W. An in vitro model of
gluten-sensitive enteropathy. Effect of gliadin on intestinal epithelial cells
of patients with gluten-sensitive enteropathy in organ culture. J Clin
Invest 1974;53:487– 500
87 Hauri HP, Kedinger M, Haffen K, Gaze H, Hadorn B, Hekkens W.
Re-evaluation of the technique of organ culture for studying gluten
toxicity in coeliac disease. Gut 1978;19:1090–8
88 Fluge G, Aksnes L. Morphological and morphometric assessment of
human duodenal biopsies maintained in organ culture. In vitro
influences of gluten in coeliac disease. Scand J Gastroenterol
1981;16:555–67
89 Bonamico M, Sabbatella L, Di Tola M, Vetrano S, Ferri M, Nenna R,
Mariani P, Picarelli A. Antiendomysial antibody detection in biopsy
culture allows avoidance of gluten challenge in celiac children. J Pediatr
Gastroenterol Nutr 2005;40:165– 9
90 Picarelli A, Di Tola M, Sabbatella L, Anania MC, Calabro A, Renzi D, Bai
JC, Sugai E, Carroccio A, Di Prima L, Bardella MT, Barisani D,
Ribes-Koninckx C, Aliaga ED, Gasparin M, Bravi E; Multicentre Organ
Culture System Study Group. Usefulness of the organ culture system in
the in vitro diagnosis of coeliac disease: a multicentre study. Scand J
Gastroenterol 2006;41:186– 90
91 Fina D, Sarra M, Caruso R, Del Vecchio Blanco G, Pallone F, MacDonald
TT, Monteleone G. Interleukin 21 contributes to the mucosal T helper cell
type 1 response in coeliac disease. Gut 2008;57:887– 92
92 Maiuri L, Ciacci C, Auricchio S, Brown V, Quaratino S, Londei M.
Interleukin 15 mediates epithelial changes in celiac disease.
Gastroenterology 2000;119:996– 1006
93 Maiuri L, Ciacci C, Raia V, Vacca L, Ricciardelli I, Raimondi F, Auricchio
S, Quaratino S, Londei M. FAS engagement drives apoptosis of
enterocytes of coeliac patients. Gut 2001;48:418– 24
94 Zanzi D, Stefanile R, Santagata S, Iaffaldano L, Iaquinto G, Giardullo N,
Lania G, Vigliano I, Vera AR, Ferrara K, Auricchio S, Troncone R,
Mazzarella G. IL-15 interferes with suppressive activity of intestinal
regulatory T cells expanded in celiac disease. Am J Gastroenterol
2011;106:130817
95 Bernardo D, Garrote JA, Allegretti Y, Leo
´nA,Go
´mez E, Bermejo-Martin
JF, Calvo C, Riestra S, Ferna
´ndez-Salazar L, Blanco-Quiro
´s A, Chirdo F,
Arranz E. Higher constitutive IL15R alpha expression and lower IL-15
response threshold in coeliac disease patients. Clin Exp Immunol
2008;154:64–73
96 Benahmed M, Meresse B, Arnulf B, Barbe U, Mention JJ, Verkarre V,
Allez M, Cellier C, Hermine O, Cerf-Bensussan N. Inhibition of TGF-beta
signaling by IL-15: a new role for IL-15 in the loss of immune
homeostasis in celiac disease. Gastroenterology 2007;132:994– 1008
97 Matysiak-Budnik T, Candalh C, Cellier C, Dugave C, Namane A,
Vidal-Martinez T, Cerf-Bensussan N, Heyman M. Limited efficiency of
prolyl-endopeptidase in the detoxification of gliadin peptides in celiac
disease. Gastroenterology 2005;129:786– 96
(Received September 1, 2011, Accepted October 19, 2011)
.................................. ..... ..... ..... ..... ..... ...... .............................. ..... ...... ..... ..... ..... ..... ..... ..... ...... ..
Lindfors et al. In vitro models for gluten toxicity 125
... Gluten is a protein present in wheat, barley, rye, and occasionally in other grains, that is responsible for the elasticity and pliability of dough, making it an essential ingredient in effect of gliadin-derived peptides. It has been extensively utilized in previous GRD-related studies to help determine the pathogenesis of these diseases [48][49][50]. ...
Article
Full-text available
Background: Non-celiac wheat sensitivity (NCWS) is a poorly understood gluten-related disorder (GRD) and its prominent symptoms can be ameliorated by gluten avoidance. This study aimed to determine the effectiveness of a probiotic mixture in hydrolyzing gliadin peptides (toxic components of gluten) and suppressing gliadin-induced inflammatory responses in Caco-2 cells. Methods: Wheat dough was fermented with a probiotic mix for 0, 2, 4, and 6 h. The effect of the probiotic mix on gliadin degradation was monitored by SDS-PAGE. The expression levels of IL-6, IL-17A, INF-γ, IL-10, and TGF-β were evaluated using ELISA and qRT-PCR methods. Results: According to our findings, fermenting wheat dough with a mix of B. longum, L. acidophilus, and L. plantarum for 6 h was effective in gliadin degradation. This process also reduced levels of IL-6 (p = 0.004), IL-17A (p = 0.004), and IFN-γ (p = 0.01) mRNA, as well as decreased IL-6 (p = 0.006) and IFN-γ (p = 0.0009) protein secretion. 4 h fermentation led to a significant decrease in IL-17A (p = 0.001) and IFN-γ (p = 0.003) mRNA, as well as reduced levels of IL-6 (p = 0.002) and IFN-γ (p < 0.0001) protein secretion. This process was also observed to increase the expression levels of IL-10 (p < 0.0001) and TGF-β (p < 0.0001) mRNA. Conclusions: 4 h fermentation of wheat flour with the proposed probiotic mix might be a good strategy to develop an affordable gluten-free wheat dough for NCWS and probably other GRD patients.
... The in vitro models of CD include epithelial cell culture model (Barone et al. 2017), T cell model (Stoven, Murray, and Marietta 2013;Sulic et al. 2015), small intestinal mucosal biopsy organ culture (Lindfors et al. 2012;Sulic et al. 2015), and the combination of human-induced pluripotent stem cells (hiPSC) with organ chip technology (Moerkens et al. 2019). In vitro model can simulate the interaction between cells, but ignore the interaction between cells and the internal environment, which cannot restore the practical situation of CD patients. ...
Article
Full-text available
Celiac disease (CD) is an autoimmune intestinal disease caused by intolerance of genetically susceptible individuals after intake of gluten-containing grains (including wheat, barley, etc.) and their products. Currently, CD, with "iceberg" characteristics, affects a large population and is distributed over a wide range of individuals. This present review summarizes the latest research progress on the relationship between CD and gluten. Furthermore, the structure and function of gluten peptides related to CD, gluten detection methods, the effects of processing on gluten and gluten-free diets are emphatically reviewed. In addition, the current limitations in CD research are also discussed. The present work facilitates a comprehensive understanding of CD as well as gluten, which can provide a theoretical reference for future research.
... In particular, the reduced efficacy of Nrf2-activated cyto-protections has been associated with the pathogenesis of celiac disease (CD) (Patlevič et al., 2016;Sido et al., 1998), an autoimmune enteropathy caused by the ingestion of vegetal proteins (wheat prolamin/gliadin and glutenins) in genetically predisposed individuals. Surprisingly, despite the growing worldwide prevalence of CD (Tack et al., 2010), only a limited number of in vitro models are available to date (Stoven et al., 2013) and -among themthe human colon adenocarcinoma cell line (Caco-2) is the most widely used in vitro model (Lindfors et al., 2012). These cells undergo a spontaneous differentiation leading to the formation of a cell monolayer expressing protein profiles partially resembling those of the human small intestine (Lenaerts et al., 2007). ...
Article
The resemblance of physiological traits of cell lines with their target/original tissue is an important prerequisite for the choice of the in vitro model. Although cytoprotective defenses, activated by the nuclear factor erythroid 2-related factor2 (Nrf2), have a preeminent importance in intestinal protection, nevertheless their levels in in vitro models have been never compared with those of their original tissue. Basal level of Nrf2-mediated defenses in murine enterocyte cells (Mode-K) and in human colon adenocarcinoma cells -at differentiated (DCaco2) or confluent stage (CCaco2)- were compared with those found in mouse or human duodenum. The pro-oxidant and cytotoxic effects of peptic-tryptic digest of gluten prepared from wheat bread (PT-b), einkorn (PT-e) or durum wheat (PT-d) were evaluated in Mode-k and DCaco2 by combining enzymatic, immune-enzymatic and real-time PCR assay. The results presented reveal that Mode-k cells resemble cytoprotective defenses of human/murine duodenum and are more susceptible to pro-oxidant, cytotoxic and pro-inflammatory effect of gliadin digest (in comparison with Caco2). Prolamins digests from the considered wheat exhibit different inhibitory effect on Nrf2-mediated defenses (PT-b > PT-e > PT-d). These data indicate, for the first time, that Mode-k are a reliable model for investigating wheat prolamins toxicity and for evaluating the signaling pathway of gluten-associated disease.
... Although the gold standard for the study of treatment of CD involves the use of in vitro organ culture of specimens of intestinal mucosa obtained from celiac patients, the use of Caco-2 cells as in vitro model of CD has some advantages. (1) It makes it possible to discriminate the contribution of each of the two branches of immunity to the overall IFN-γ-or gliadin-dependent response, (2) It is inexpensive, (3) The Caco-2 cells may also be used to study in vitro the preventive effects of bioavailable dietary bioactive compounds toward celiac inflammation, and (4) Caco-2 cells are used more extensively than T84 cells in CD research, notably for testing of novel treatments (Lindfors, Rauhavirta, Stenman, Maki, & Kaukinen, 2012). The disadvantages of the Caco-2 cell line include the inability to reproduce all the characteristics of a human intestinal epithelium and the transport of lipophilic molecules which may be decreased since Caco-2 cells do not contain a mucus layer with bile acids as human intestinal cells do (Lea, 2015). ...
Article
The objective of this research was to investigate the efficacy of procyanidin B2 rich cocoa extracts on interferon-gamma (IFN-γ) or gliadin peptide p31-43-induced transglutaminase-2 and interleukin-15 (IL-15) in Caco-2 cell model of in vitro celiac disease. Cysteamine was used as a positive control inhibitor of TG2. Procyanidin B2-rich extract reduced IFN-γ- and gliadin p31-43-induced TG2 activity by 77% and 45%, respectively. Cocoa extract containing 4.9–289 µg/mL of procyanidin B2 dose-dependently reduced IFN-γ-induced IL-15 secretion to the level of IL-15 in control cells. Similarly, cocoa extract containing 4.9–289 µg/mL of procyanidin B2 dose-dependently reduced gliadin p31-43-induced IL-15 to the level in control cells. Procyanidin B2-rich cocoa extracts reduced the activities of other inflammatory biomarkers including COX-2, IL-1β, IL-6, and IL-8 in both IFN-γ and p31-43-treated Caco-2 cells. Caffeine or theobromine, at the concentration found in the cocoa extracts, did not contribute to the activity of procyanidin B2 against TG2 or IL-15.
... Ex-vivo gluten application on gluten-free diet intestinal biopsy, bypass the obstacle. Many ex-vivo, in vitro models were described, using human originated intestinal cell-lines or CD patients small bowel mucosal biopsies [41,42]. We know nowadays that gluten or gliadin, when incubated with primary cultures of CD patients on glutenfree diet,induces morphological as well as immunological aberrations. ...
Article
Full-text available
Celiac disease is a multi-organ disorder which is highly variable in its clinical expression, presenting multiple enteric and extraintestinal manifestations. However, most of treating physicians and dieticians regard celiac disease as a chronic, gradually evolving, asymptomatic or hypo-symptomatic entity. The present mini-review aims to screen the literature for acute presentations of celiac disease and to increase the awareness of the medical communities, for such a possibility. It appears that the disease can present acutely in multiple symptomatic, phenotypic and laboratory pictures. The acute presentation involves mainly the gastrointestinal tract and adjacentorgans like liver and gallbladder, however, extraintestinal and remote organs presentations can occur.
... However, gliadin may increase intestinal permeability through the subsequent attraction of macrophages in the intestinal submucosa. 9 Celiac disease is a life-long autoimmune disorder, occurring 1 in 100 individuals worldwide. On the epidemiological basis, India has a promising number of gluten sensitive patients in North Indian population. ...
Article
Gluten-related diseases such as wheat allergy, celiac disease, and gluten intolerance are widespread in the globe to genetically pre-disposed individuals. The present study aims to develop wheat-gluten induced BALB/c murine model for addressing wheat-gluten related disorders by sensitizing the wheat gluten through the route of intraperitoneal and oral challenge in prolonged days. During the sensitization, the sera were collected for specific anti-gliadin antibodies response and pro-inflammatory markers quantification. An ex-vivo primary cells and organs were collected for subsequent analysis of inflammatory profile. Prolonging sensitization of gluten can moderate the antigen-specific inflammatory markers such as IL-1β, IL-4, IL-15, IL-6, IFN-ϒ and TNF-α level in mice sera. However, ex-vivo primary cells of splenocytes (SPLs) & intestinal epithelial lymphocytes (IELs) significantly increased the IL-6, IL-15, IL-1β, & IL-4 levels in G + (gliadin and gluten) treated cells. Histopathology staining of jejunum sections indicates that enterocyte degeneration in the apical part of villi and damage of tight junctions in G + (gliadin and gluten) sensitised murine model. Immunohistochemistry of embedded jejunum sections showed significant expression of positive cells of IL-15, tTG and IL-4 in G + sensitised murine model. In contrast, all markers of gluten-related disorders are expressed exclusively such as tTG, ZO-1, IL-15, IL-6, IL-4 and intestinal inflammation was mediated by iNOS, COX-2, TLR-4 and NF-kBp50 signalling mechanism in G+ sensitized mice.
... The Caco-2 cell line was purchased from Iran Pasteur Institute (Tehran, Iran) and were cultured in RPMI 1640 supplemented with 10% FBS (Gibco, Manchester, UK) at 37°C in a humid incubator with 5% CO 2 . The cells were cultured in the presence of different concentration of gluten for 24 h as previously reported [14]. Then, RNA was isolated using a RNA extraction kit (Bioflux, Basel, Switzerland) and RNA was transcripted into cDNA using of Bioneerkit (Bioneer, Daejeon, South Korea). ...
Article
Full-text available
Accumulating evidence show that many inflammatory cytokines are involved in pathophysiology of celiac disease (CD). CCL28 known as mucosa associate epithelial chemokine (MEC) is produced by mucosa and chemoattracts IgA-producing B cells into the mucosa. However, its levels in patients with CD have not yet been elucidated. CCL28 levels and anti-tTTG (IgA) were detected in the serum of 28 new cases of CD, 32 cases of treated patents and 32 normal individuals by Elisa. Moreover, the effect of gluten on intestinal cells, Caco-2, was examined by RT-PCR. Our data show that (i) the levels of CCL28 is significantly higher in patients with CD than normal individuals, (ii) CCL28 levels is reduced in patients with CD who had gluten-free diets. Accordingly, we observed that CCL28 expression is upregulated in a dose-dependent manner when the Caco-2 cells were cultured in the presence of gluten. In conclusion, gluten enhances CCL28 expression and that CCL28 could be a novel biomarker for diagnosis and following up the patients with CD. However, further investigation in a larger number of patients is required.
Article
Full-text available
Scope Proteolysis-resistant gliadin peptides are intensely investigated in biomedical research related to Celiac Disease and gluten-related disorders. Herein, the first integrated supramolecular investigation of pepsin-digested gliadin peptides, p-gliadin, is presented in combination with its functional behavior in Caco-2 cell line. Methods and results First, we investigated gliadin degradation by pepsin at pH 3, and the physicochemical properties of p-gliadin were compared with gliadin. An integrated approach using interfacial, spectroscopic, and microscopic techniques revealed that the p-gliadin forms spontaneously soluble large supramolecular structures, mainly oligomers and fibrils capable of binding amyloid-sensitive dyes. The self-assembly of p-gliadin starts at a concentration of 0.40 μg/ml. Second, we stimulated CaCo-2 cells with the p-gliadin supramolecular system and screened the mRNA expression levels of a panel of genes involved in cellular inflammation, apoptosis, permeability, and chemoattraction of immune cells. Our findings suggest that p-gliadin composed of supramolecular structures triggers significant mRNA up-regulation (p < 0.05) of pro-apoptotic biomarkers (ratio Bcl2/Bak-1), chemokines (CCL2, CCL3, CCL4, CCL5, CXCL8) and the chemokine receptor CXCR3. Conclusions This work demonstrates that p-gliadin is interfacial active, forming spontaneously amyloid-type structures that trigger genes in the Caco-2 cell line involved in the recruitment of specialized immune cells. This article is protected by copyright. All rights reserved
Article
Since the 1970s, the role of infectious diseases in the pathogenesis of Graves' disease (GD) has been an object of intensive research. The last decade has witnessed many studies on Yersinia enterocolitica, Helicobacter pylori and other bacterial organisms and their potential impact on GD. Retrospective, prospective and molecular binding studies have been performed with contrary outcomes. Until now it is not clear whether bacterial infections can trigger autoimmune thyroid disease. Common risk factors for GD (gender, smoking, stress, and pregnancy) reveal profound changes in the bacterial communities of the gut compared to that of healthy controls but a pathogenetic link between GD and dysbiosis has not yet been fully elucidated. Conventional bacterial culture, in vitro models, next generation and high-throughput DNA sequencing are applicable methods to assess the impact of bacteria in disease onset and development. Further studies on the involvement of bacteria in GD are needed and may contribute to the understanding of pathogenetic processes. This review will examine available evidence on the subject.
Article
Full-text available
Under physiological conditions the gut-associated lymphoid tissues not only prevent the induction of a local inflammatory immune response, but also induce systemic tolerance to fed antigens. A notable exception is coeliac disease, where genetically susceptible individuals expressing human leukocyte antigen (HLA) HLA-DQ2 or HLA-DQ8 molecules develop inflammatory T-cell and antibody responses against dietary gluten, a protein present in wheat. The mechanisms underlying this dysregulated mucosal immune response to a soluble antigen have not been identified. Retinoic acid, a metabolite of vitamin A, has been shown to have a critical role in the induction of intestinal regulatory responses. Here we find in mice that in conjunction with IL-15, a cytokine greatly upregulated in the gut of coeliac disease patients, retinoic acid rapidly activates dendritic cells to induce JNK (also known as MAPK8) phosphorylation and release the proinflammatory cytokines IL-12p70 and IL-23. As a result, in a stressed intestinal environment, retinoic acid acted as an adjuvant that promoted rather than prevented inflammatory cellular and humoral responses to fed antigen. Altogether, these findings reveal an unexpected role for retinoic acid and IL-15 in the abrogation of tolerance to dietary antigens.
Article
The human colon carcinoma cell line Caco-2 grown in vitro under standard culture conditions in the absence of inducers of differentiation spontaneously exhibits signs of structural and functional differentiation and polarization. The differentiation is total at late confluency. Transmission and scanning electron microscopy show that the entire upper side of the monolayer is covered with typical brush border microvilli which extend perpendicularly to the surface. The asymmetry of the cells and the presence of tight junctions suggest that the monolayer is polarized. Functionally the differentiation is characterized by high levels of activity of the brush border associated enzymes. The levels of alkaline phosphatase and sucrase-isomaltase are nearly 50% of those found in similar preparations of human small intestine, and 10% in the case of aminopeptidase. The presence of the enzymes is further confirmed by the immunofluorescence staining of the cells with anti-sucrase-isomaltase antibodies. The functional enterocytic differentiation of the brush border microvilli is associated with the formation of domes which are typical of transporting epithelial monolayers. In the early proliferation phase of the culture 70% of the cells are structurally differentiated, but only a few of them are immunoreactive with anti-sucrase-isomaltase antibodies, and the enzyme activities are low.
Article
Background: Coeliac disease (CD) is an enteropathy mediated by gluten specific T cells which secrete interferon γ (IFN-γ) when stimulated by gluten peptides presented by HLA-DQ2 or DQ8 molecules. Residues 62–75 of α2 gliadin have been proposed as the immunodominant epitope in the majority of CD patients. Deamidation by tissue transglutaminase (tTG) of the glutamine (Q) at position 65 to glutamic acid (E) is essential for T cell stimulation. Aims: To investigate the antigenicity of this peptide and to establish whether its T cell activating properties can be downregulated by the formation of altered peptide ligands. Patients: Individuals with known CD. Methods: Peptide G4 corresponding to α2 gliadin residues 62–75, Q-E65 and analogues, substituting each amino acid, except E65, in turn for alanine residues, were synthesised. Small intestinal biopsies were obtained from patients. Biopsies were cultured overnight with a peptic/tryptic digest of gliadin (PTG). Lymphocytes were cultured and restimulated with tTG treated PTG. A T cell line was cloned and clones tested for stimulation and IFN-γ production in response to G4 and its analogues. Results: Some high activity clones were isolated with, for example, a stimulation index (SI) of 15 to G4 and secreting 327 pg/ml of IFN-γ. Substitution of amino acids at several positions abolished or downregulated stimulation and IFN-γ production. Conclusions: Peptide G4 is highly immunogenic. Certain amino acid substitutions in peptide G4 abolish T cell reactivity while others are partial agonists which may have potential in immunomodulation in this condition.
Article
BACKGROUND—Villus atrophy is the most distinctive sign of untreated coeliac disease (CD) and epithelial apoptosis is considered to be involved in this stage of the coeliac lesion. The extent of villus atrophy is, however, not homogeneous and patients with patchy or mild lesions have been described. AIMS—To address: (a) the degree of "patchiness" in untreated CD patients; and (b) to clarify if apoptosis, and eventually which trigger drives it, causes epithelial damage. PATIENTS—Twenty of 40 untreated, 14 treated coeliac patients, and 15 controls received five or more multiple duodenal biopsies; the remaining 20 untreated CD patients had no more than three biopsies. METHODS—All biopsies were analysed to monitor the presence of a "flat" mucosa. Biopsies of 14 untreated, 10 treated coeliacs, and seven controls were cultured with or without gliadin. DNA fragmentation was studied by terminal deoxynucleotidyl transferase (TdT) mediated dUTP digoxigenin nick end labelling (TUNEL), and FAS and Ki67 expression by immunohistochemistry. Antiendomysium antibodies (EMA) were surveyed in biopsy culture supernatants. RESULTS—A pattern of patchy duodenal lesions was observed in all untreated CD patients biopsied up to five times. High enterocyte FAS expression, and a high number of TUNEL+ and Ki67+ enterocytes were detected in areas with villus atrophy but not in those with a normal morphology (p 0.01) while agonist anti-FAS monoclonal antibody increased it (p<0.001). CONCLUSIONS—Patchy lesions are observed in untreated CD mucosa and epithelial FAS engagement is a key trigger in driving villus atrophy in CD. Keywords: apoptosis; FAS; enterocytes; coeliac disease
Article
Background & Aims: Celiac disease is caused by T-cell responses to wheat gluten-derived peptides. The presence of such peptides in other widely consumed grains, however, has hardly been studied. Methods: We have performed homology searches to identify regions with sequence similarity to T-cell stimulatory gluten peptides in the available gluten sequences: the hordeins of barley, secalins of rye, and avenins of oats. The identified peptides were tested for T-cell stimulatory properties. Results: With 1 exception, no identical matches with T-cell stimulatory gluten peptides were found in the other grains. However, less stringent searches identified 11 homologous sequences in hordeins, secalins, and avenins located in regions similar to those in the original gluten proteins. Seven of these 11 peptides were recognized by gluten-specific T-cell lines and/or clones from patients with celiac disease. Comparison of T-cell stimulatory sequences with homologous but non-T-cell stimulatory sequences indicated key amino acids that on substitution either completely or partially abrogated the T-cell stimulatory activity of the gluten peptides. Finally, we show that single nucleotide substitutions in gluten genes will suffice to induce these effects. Conclusions: These results show that the disease-inducing properties of barley and rye can in part be explained by T-cell cross-reactivity against gluten-, secalin-, and hordein-derived peptides. Moreover, the results provide a first step toward a rational strategy for gluten detoxification via targeted mutagenesis at the genetic level.