You are on page 1of 25

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

net/publication/356538001

Celiac Disease

Chapter · January 2022


DOI: 10.1007/978-3-030-80068-0_40

CITATIONS READS

2 2,365

2 authors:

Stefano Guandalini Valentina Discepolo


The University of Chicago Medical Center University of Naples Federico II
280 PUBLICATIONS   14,296 CITATIONS    83 PUBLICATIONS   1,551 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

probiotics View project

Natural history of celiac disease in a Potential celiac patients cohort View project

All content following this page was uploaded by Stefano Guandalini on 05 February 2022.

The user has requested enhancement of the downloaded file.


Celiac Disease
40
Stefano Guandalini and Valentina Discepolo

Introduction men appear to underutilize health-care services [12, 13].


Over the past several years, it has also become clear that CD
Celiac disease (CD) is a complex autoimmune disorder elic- prevalence has been progressively increasing [9, 14–18].
ited by the ingestion of gluten (the main storage protein in While the availability of reliable serological tests to screen
wheat, barley, and rye) in genetically predisposed individu- for CD as well as an increased awareness, especially in
als expressing specific human leukocyte antigen (HLA)- North America, can be considered responsible for increased
class II haplotypes (called DQ2 and DQ8) and causing an diagnostic rates, reliable epidemiological data convincingly
inflammatory disorder of the small intestine. CD is charac- show worldwide a true increase in prevalence, of the order
terized by a variable combination of elevated titers of celiac-­ of doubling rates every 20 years or so. In Northern Sweden,
specific autoantibodies, an inflammatory enteropathy with an epidemiological investigation employing a combined
variable degrees of severity, and a gastrointestinal (GI) as serological/endoscopic approach in an unselected popula-
well as a wide range of extra intestinal complaints [1] whose tion of 1000 adults found a prevalence of almost 2% [19].
severity varies from asymptomatic to life-threatening. CD is Since obviously genetic changes cannot be blamed for this
well known to be often associated with other autoimmune rapid change, a variety of environmental factors have been
conditions such as type 1 diabetes and autoimmune thyroid- called as potentially responsible for such increase, but their
itis [2], as well as with some congenital disorders such as respective role is still unclear [20]. In fact, for some sus-
Down [3, 4] Turner [5], and most recently also Williams-­ pected factors the evidence is at best equivocal or contradic-
Beuren syndrome [6]. tory, among them newer varieties of wheat obtained by
breeding techniques [21–23], seasonality and modalities of
delivery [24–28], and exposure to antibiotics [29–33]. On
 pidemiology
E the other hand, infections in early life [34, 35] and espe-
cially respiratory infections [36] including influenza [37]
Currently, it is estimated that CD has a global prevalence and infections by Enterovirus [38] appear likely to increase
around 1% of the general population [7], although there are the risk of CD development. In this respect, the finding of
important variations between different geographical areas the role of reovirus in inducing inflammatory response to
[8–10], even within a single country [11]. Furthermore, CD dietary antigens and favoring CD development [39] appears
is more prevalent in females than males [7]. However, since a key finding. Interestingly, vaccinations do not represent a
studies of prevalence based on screening do not show such risk factor [40].
clear-cut difference, it is currently thought that the female Infant feeding practices have also been deeply scrutinized
predominance could also in part be attributed to the fact that in a number of observational and interventional studies, and
their results can currently be summarized as follows [41, 42]:

S. Guandalini
• Breast feeding, long thought to have a protective effect on
Department of Pediatrics, Section of Gastroenterology, Hepatology
and Nutrition, University of Chicago, Chicago, IL, USA the development of CD, appears not to be beneficial in
e-mail: sguandalini@peds.bsd.uchicago.edu this regard [43–46].
V. Discepolo (*) • Avoidance of cow’s milk in early life (suggested to reduce
Department of Translational Medical Sciences, Section of the risk of CD later in life in children at genetic risk) has
Pediatrics, University of Naples Federico II, Naples, Italy no protective effect [47].
e-mail: valentina.discepolo@unina.it

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 525
S. Guandalini, A. Dhawan (eds.), Textbook of Pediatric Gastroenterology, Hepatology and Nutrition,
https://doi.org/10.1007/978-3-030-80068-0_40
526 S. Guandalini and V. Discepolo

• Early (before the end of the third month) introduction Genetic Factors
of gluten is not recommended as it is considered a risk
factor [48]. In contrast to this, however, a recent pro- The weight of genetic predisposing factors in CD pathogen-
spective study in the UK [49] showed a different sce- esis is relevant, as initially suggested by the observation of a
nario, suggesting on the contrary that – similar to some strong familiar aggregation [53], with a prevalence of CD
food allergies  – early introduction of gluten could be among first-degree relatives of around 10%. The most impor-
associated with a reduced rate of subsequent CD. As it tant and best-characterized susceptibility gene is the HLA
can be seen therefore, the field is still a rapidly evolv- class II; in particular, the HLA-DQ2 and HLA-DQ8 alleles
ing one. are required for CD development. The concordance rate is
• Introducing gluten in small amounts [44] or after the first about 30% among HLA-identical siblings, 80% in monozy-
year of life [43] does not offer protection, as the strongest gotic twins, and 10% in dizygotic twins [54]. The HLA-DQ2
factors favoring the onset of CD appear to be gender and molecules are expressed on the surface of antigen-presenting
HLA status. cells and contain positively charged pockets that preferen-
• Intake of large amounts of gluten during the first 2 years tially bind negatively charged epitopes, such as deamidated
of age increases the risk of CD in genetically susceptible gliadin peptides (DGP), and present them to cluster of dif-
children [45]. ferentiation (CD)-4+ T cells, thereby activating them [55,
• The possible role of maternal diet during pregnancy and 56].
on diet of the child after weaning in influencing the onset
of CD has also been investigated. A recent study on more  he Central Role of HLA-DQ Haplotype
T
than 80,000 participants [50] showed that higher maternal HLA genes are located on chromosome 6 and are divided
fiber intake (median 29.5  g/day) was associated with a into three classes (I–III). HLA-DQ (6p21.3) belongs to class
lower risk of CD in the offspring, and that gluten intake II and is composed of a heterodimer located on antigen-­
during pregnancy (median 13.0 g/d) was associated with presenting cells and encoded by HLA-DQA1 and HLA-­
a higher risk of childhood CD, suggesting that maternal DQB1. Each of the copies of the HLA-DQ gene encodes for
diet during pregnancy could impact later CD autoimmu- a heterodimer, resulting in four proteins (one couple per
nity [50]. chromosome). HLA-DQ2 homozygote (DQB1*02 on both
• In a large prospective study of dietary patterns of young chromosomes) subjects have the highest risk to develop CD,
children, Barroso et al. [51] found that high consumption fivefold higher than heterozygotes [57, 58]. Even if rarer in
of vegetables and grains and low consumption of refined the general population, DQ2 homozygosis is characteristic
cereals and sweet beverages are associated with lower of 25% of all CD patients and often relates to a more severe
odds of developing CD autoimmunity, the first step in the clinical outcome, as suggested by its association with earlier
development of the disease. Thus, an eating pattern that disease onset [59, 60] and its higher prevalence in patients
can be reasonably liked to a Mediterranean diet, as with refractory CD [61], a rare but severe complication of
opposed to the “Western” diet that appears to be linked CD.  As a matter of fact, the immune response arising in
with various chronic inflammatory disorders, may have a homozygous individuals is stronger than the response in het-
protective effect. erozygous individuals [53, 62]. The most common configu-
ration (more than 50% of CD patients) is represented by
A plausible mediator of many of the environmental fac- DQ2.5 (DQB1*02/DQA1*05) heterozygotes [58].
tors mentioned is therefore the microbiome, whose composi- HLA-DQ8 is found in 5–10% of CD patients [58, 63], with
tion and modifications have been shown to play a critical role HLA-DQ8 homozygosis conferring increased risk compared
in the onset (or prevention) of allergic and inflammatory dis- to HLA-DQ8 heterozygosis [64]. It must be noted that about
orders. In recent years there has been therefore an increasing 5% of CD patients carry only one of the HLA-DQ2 heterodi-
focus on the role of microbiota in the development of CD mer alleles (HLA-DQA1*05 or HLA-DQB1*02), therefore
[52] (see below). encoding for the so-called half heterodimer. These patients
constitute the overwhelming majority of CD subjects not
carrying the full HLA-DQ2 and/or -DQ8 alleles [65]. The
Etiopathogenesis highest risk group includes individuals who inherit both
DQ8 and DQ2. Only less than 1% of patients who fulfill
Like most multifactorial disorders, CD is the result of a com- clinical criteria for CD do not carry the DQ2 (including half
plex interaction between genes and immune status of the heterodimer) nor the DQ8 alleles [65]. Thus, in the clinical
host and environmental triggers. practice, CD can be virtually excluded in individuals lacking
40  Celiac Disease 527

HLA-DQ2 or HLA-DQ8. Interestingly, 30% of European miological and genetic studies. GWAS identified
descent individuals carry HLA-DQ2 susceptibility allele; polymorphisms in genes involved in the response to viral
however, only about 4% of these individuals will develop infections to be associated with CD. Higher rates of summer
CD [57]. This suggests that even though those molecules are births were described in children with CD, suggesting that
necessary, they are not sufficient to induce the disease, exposure of 4–6-month-old infants to winter-linked viral
implying that other genetic and environmental factors must infections such as rotavirus may play a role. The first correla-
contribute to it. tion between viruses and CD dates back to the 1980s when a
homology between alpha gliadin and a protein of the human
 on-HLA Genetic Susceptibility Factors
N adenovirus [70] had been described and related to increased
HLA alleles explain 35% of the disease genetic susceptibil- frequency of adenovirus infection in CD patients [71] com-
ity, while the remaining is due to numerous non-HLA genes pared to controls. Increased anti-rotavirus antibody titers have
that singularly contribute to a much lower extent to CD heri- also been associated with a moderate, but significantly
tability. Genome-wide association studies (GWAS) per- increased risk of CD in HLA-susceptible children [72]; subse-
formed over the years identified 40 CD-associated quently [73] anti-rotavirus antibodies have been found related
susceptibility loci (including the HLA locus) [66]. More with CD but not with type 1 diabetes mellitus (T1DM) onset,
recently a meta-analysis identified 2 additional non-HLA despite the common genetic background and similar patho-
CD risk loci reaching genome-wide significance, bringing genic mechanisms. Over the past decade, several publications
the number up to 41 [67]. Notably, 64% of the non-HLA loci have explored the association between viral infections and CD
were shared with at least another autoimmune disease (e.g., development. Overall, they reported that a high number of
type 1 diabetes), reinforcing the idea that autoimmune disor- infections in the first months of life are associated with an
ders may share common pathogenic pathways [68]. In those increased risk of later CD development [74]. Of note, the risk
regions, more than 115 non-HLA genes have been associated increased synergistically if, in addition to infectious episodes,
to CD, individually contributing only modestly to the overall dietary gluten was first introduced in large amounts after
disease risk. Twenty-eight of them encode for molecules breastfeeding was discontinued [35]. Respiratory infections in
involved in the immune response, reinforcing the central role particular [37, 75] appear to be the ones most commonly asso-
of immune dysregulation in CD pathogenesis. Post-GWAS ciated with CD.  Importantly, the risk variance explained by
will need to focus on elucidating the functional basis of these respiratory infections was higher than that explained by sex or
genetic variants and in particular the role of regulatory varia- HLA [36]. And recently, a clear role for Enterovirus infections
tion. Furthermore, considering multiple gene variants may in early life has also been reported [38]. Altogether these
help refine risk prediction models and identify high-risk indi- observations provide a strong support for the role of viral
viduals that could benefit from preventive strategies. infections in CD development. Nevertheless, despite viral
infections being very common and HLA-DQ2/8 being present
in about 40% of the general population, only 1% develops CD,
Environmental Factors raising questions about the pathogenic mechanisms behind
this association. The first work showing evidence for a mecha-
The high impact of genetic predisposing factors in CD does nism behind the association of viral infections and CD was
not exclude a key role of the environment in triggering dis- published in 2017 and showed that a specific strain of reovirus
ease development. In fact, as mentioned above, the increased (T1L), despite being cleared by the host, could disrupt immune
incidence of CD in the past decades cannot be explained by homeostasis at intestinal sites of oral tolerance by suppressing
genetic drift and most likely results from faster changes in peripheral regulatory T cell conversion and promoting a proin-
the environment. In an elegant review, Abadie et  al. [69] flammatory T helper cell response to orally ingested gluten
showed that CD prevalence does not perfectly correlate, as it [39]. Importantly, the two responses depended on distinct but
would be predicted, with the levels of wheat consumption interplaying pathways. Lastly this study showed increased
and frequencies of HLA-DQ predisposing alleles, thus anti-reovirus antibodies in CD patients, suggesting that the
emphasizing the role of environmental factors in CD onset. risk of later disease development is independent from the
The role of infant feeding practices on CD development severity or virulence of the infection; in fact reovirus causes
has been discussed above. Below, we will discuss the role of asymptomatic infections in humans. Whether there is an early
viral infections and intestinal microbiota. time window during which infections are more likely to pro-
mote CD, remains to be determined, as well as the interplay
Viral Infections with other factors such as timing and amount of first dietary
Infective agents have been investigated as environmental fac- gluten introduction, breastfeeding, and declining of maternal
tors that contribute to trigger CD, as suggested by both epide- antibody titers.
528 S. Guandalini and V. Discepolo

Finally, the role of viral infections beyond promoting loss the adaptive arms of the immune response [83]. In this
of tolerance to gluten, such as their ability to enhance the model, two hits are required, each one being necessary but
progression of the disease to tissue damage, is unexplored. not sufficient for the development of CD: one is the activa-
tion of stress markers in intestinal epithelial cells and the
Intestinal Microbiota second is the rise of a gluten-specific proinflammatory CD4+
The composition of intestinal microbiota as well as bacte- T cell response in the lamina propria. These two events lead
rial metabolic products can affect intestinal epithelium to the full activation of lymphocytes in the intestinal epithe-
function and mucosal immune homeostasis. Both quantita- lium (intraepithelial lymphocytes (IELs)), final responsible
tive and qualitative differences in the composition of the of tissue destruction [84, 85].The major players of the patho-
intestinal microbiome have been observed in patients with genic events leading to full-blown CD are illustrated in the
autoimmune disorders [76, 77], as well as CD, as recently following sections.
reviewed [52] when compared to healthy individuals. In
addition, disease activity and response to the diet may also Gluten
play a role [78]. The immunological reaction occurring in CD patients fol-
The variability between fecal and duodenal microbial lows the ingestion of gluten-containing cereals: wheat, rye,
niches, the different methodologies used across studies, and and barley. Glutenins and gliadins, typical gluten compo-
the heterogeneity of CD patients make the identification of a nents, are responsible for the viscosity and elasticity of the
CD-specific microbiota a hard task. Nevertheless, most evi- wheat dough [86]. Their high concentration of glutamine and
dences point to an expansion of Proteobacteria and opportu- proline residues (35% and 15% of the total amino acid con-
nistic pathogens such as Neisseria [79] or Escherichia coli, as tent) renders them highly resistant to gastrointestinal
well as higher bacterial virulent genes, supporting the idea of enzymes. Indeed, the lack of prolyl-endopeptidase activity in
a shift toward a proinflammatory microbial community [78]. any of the human digestive enzymes prevents enzymatic
Even when looking at children at higher genetic risk to attack of proline-rich domains in gluten proteins. Thus, at the
develop CD, an increase in pathogenic bacteria has been end of a normal, full digestive process of gluten, many glia-
observed [80]. In fact, the HLA genotype seems to influence din peptides remain undigested in the intestinal lumen. The
the early intestinal microbial composition [81]. Moreover, mechanism through which such peptides reach the intestinal
non-HLA genes associated with protection to infections or lamina propria is not entirely clear. Gliadin is able to enhance
bacterial recognition have been related to CD, suggesting gut paracellular permeability via zonulin-mediated pathway,
that a genetically determined pattern of microbial coloniza- a molecule involved in tight junction disassembly, as sug-
tion might influence the immune maturation process and gested also by genetic studies associating tight junction
thus promote later CD onset [82]. genes with CD [87]. Moreover, there is evidence that gluten
Currently, it is still unclear whether the observed changes can cross the intestinal epithelium through the transcellular
in the microbial composition are the cause or the result of the pathway once tolerance to gluten has been broken.
intestinal inflammatory process. Indeed, epithelial altera- Even though an increase in intestinal permeability has
tions, such as those described in CD, could create a specific been shown in CD, a retro-transcytosis pathway has been
environment favoring the selective colonization of some described for secretory antibodies, potentially having a role
microbial species, thus contributing to the creation of the also for gliadin transport. In fact, the transferrin receptor
microenvironmental milieu that favors the disease develop- CD71, normally expressed on the basolateral side of entero-
ment. This is clearly a rapidly evolving field, likely to bring cytes, is overexpressed on the luminal side of the intestinal
important new acquisitions in the near future. epithelium in active CD patients, leading to an apical-to-­
basal retro-transcytosis of gliadin peptides complexed with
secretory IgA [88]. This process protects gliadin fragments
Pathogenesis from degradation and facilitates their access to the intestinal
lamina propria, thereby perpetuating the inflammatory
CD is a chronic inflammatory disorder with autoimmune process.
features, characterized by a gluten-specific T cell-mediated
immune response arising in the small intestinal mucosa upon  issue Transglutaminase 2 and Autoantibodies
T
gluten ingestion. In genetically susceptible individuals, glia- In the lamina propria, tissue transglutaminase 2 (tTG), a
din peptides activate stress pathways and provide ligands for calcium-­ dependent transamidating enzyme, mediates the
innate immune receptors, leading to the release of proinflam- conversion of glutamine residues into glutamic acid,
matory mediators that then sustain the T cell response both in ­introducing negatively charged residues into gliadin peptides
the lamina propria and in the epithelium (Fig.  40.1). This that act as immunogenic epitopes binding to HLA-DQ mol-
inflammatory reaction typically involves both the innate and ecules with relatively higher affinity, thus representing a pre-
40  Celiac Disease 529

Fig. 40.1  CD pathogenesis. Celiac disease (CD) is a multifactorial dis- (green circles) to naïve CD4+ T cells (red cells). Inflammatory DCs
order induced by gluten in genetically susceptible subjects. Several (purple) enhance the rise of a gluten-specific CD4+ T cell response,
environmental factors (i.e., viral infections, alterations in microbiome characterized by the production of high levels of proinflammatory cyto-
composition, etc.) can contribute to trigger the disease by inducing epi- kines such as IFN-γ and IL-21. Furthermore, gluten-reactive CD4+ T
thelial stress in the intestinal mucosa, and the production of innate cells provide the required help to TG2-specific B cells, presenting TG2-­
immune cytokines, such as interleukin (IL)-15 and type-1 interferon gliadin complexes, in a hapten carrier-like manner, and drive TG2-­
(IFN). Those cytokines contribute to create a proinflammatory environ- specific antibody production by plasma cells (light blue cells). IL-15
ment that enhances dendritic cell (DC) activation. Undigested gliadin and IL-21 contribute to the full activation of cytotoxic CD8+ intraepi-
peptides (purple circles) reach the intestinal lamina propria where they thelial lymphocytes (IEL). An increased epithelial expression of stress
are deamidated by tissue transglutaminase 2 (TG2). Deamidated gliadin markers (i.e., MIC, HLA-E, and IL-15) is also required to enable acti-
peptides (DGP) are more suitable to be presented by HLA-DQ mole- vated CD8+ IEL to target the intestinal epithelial cells and determine
cules. Only DCs expressing the CD-associated HLA-DQ2 and/or villous atrophy in CD patients. NKG2D natural killer group 2, member
HLA-DQ8 molecules (orange) can present deamidated gliadin peptides D, MIC macrophage inhibitory cytokine

requisite for a gluten-specific T cell response [89]. In addition reactive CD4+ T cells provide the required help to tTG-­
to its crucial role in inducing post-translational modification specific B cells in a hapten carrier-like manner and drive
of gluten peptides, tTG is also the main autoantigen for tTG-specific antibody production. In fact, a very high pro-
CD. In fact, increased serum levels of anti-tTG IgA (or IgG, portion of the gut plasma cells, which expand massively in
in case of IgA deficiency) autoantibodies is a key diagnostic the lamina propria during active CD, were shown to produce
feature of CD. Their production seems to occur at the level of IgA specific for gluten or TG2 or both.
the small intestine. The most accepted hypothesis for their
formation is that the enzyme tTG cross-links itself to gluten  he Intersection of Adaptive and Innate Immune
T
during the substrate-enzyme interaction [90]. Once internal- Responses in CD
ized, those complexes are processed and gluten epitopes bind Gluten contains a large number of peptides capable of stimulat-
HLA-DQ molecules of the B cell that may allow gluten-­ ing T cells [91]. Dendritic cells present negatively charged glia-
530 S. Guandalini and V. Discepolo

din peptides through HLA-DQ2 or HLA-DQ8 molecules to sis, villous atrophy, and crypt hyperplasia. IELs represent a
naïve CD4+ T cells, thus enhancing a gluten-specific T helper 1 heterogeneous population including TcRα/β CD8 cells,
(Th1) inflammatory response, characterized by the production NK-like cells, and TcRγ/δ cells. An increase in both TcRα/β
of high levels of interferon gamma (IFN-γ) and interleukin and TcRγ/δ populations has been described in CD patients;
(IL)-21 in the small intestinal lamina propria (Fig. 40.1). The however, the role of the former has been investigated more
gluten-specific CD4+ T cell response, which contributes also to extensively than the latter in the context of CD pathogenesis.
the autoantibody’s generation, is sustained by several cyto- A permanent reshaping of the TcRγ/δ IELs compartment has
kines, including IL-15 and IL-21, that synergistically promote been recently described in CD as characterized by an expan-
gluten-mediated increase of IFN-γ production. The expression sion of gluten-sensitive and IFN-γ-producing Vδ1+ IELs that
of IL-21, enhanced by gluten, is very high in active CD patients, the exclusion of dietary gluten was insufficient to revert [98].
while it is downregulated in potential CD [92], suggesting that TcRα/β IELs are the final responsible for tissue destruc-
this cytokine plays a key role in the development of tissue dam- tion. A fully active phenotype characterized by an upregula-
age. The rise of a gluten-­specific CD4+ T cell response could be tion of activating NKRs, downregulation of inhibitory NKRs,
triggered by specific viral infections, as shown for reovirus and expression of granzyme B and perforin is typically found
T1L strain that can induce loss of tolerance to oral antigens via in active CD patients [84]. The activation of their cytotoxic
an IRF-­1-­mediated pathway [39], but also possibly by bacterial properties requires also stimuli from the epithelial compart-
antigens. Once activated, the gluten-specific T cell response ment. An increased expression of stress molecules, such as
contributes to amplify the inflammatory response; neverthe- nonclassical MHC class I molecules (i.e., MIC and HLA-E),
less, as evident in potential CD patients, the gluten-specific T has been found in intestinal epithelial cells of active CD
cell adaptive immune response alone can occur even in the patients, as part of the innate stress response induced by gluten
absence of villous atrophy, suggesting that other signals are or by other environmental triggers. Notably, MIC and HLA-E
required to induce tissue damage. are ligands for NKG2D and CD94, respectively,  activating
In addition to the immunodominant epitopes, which, as NKRs that are upregulated on IELs in active CD patients and
discussed, are presented to CD4 naïve T cells, gliadin also whose expression is enhanced by IL-15 [83–85].
contains fragments which are able to enhance epithelial Increased levels of both IL-15 and IL-21 in the duodenal
stress and induce innate immune effects [93]. The most stud- mucosa of untreated CD patients cooperatively promote the
ied fragment is the peptide 31–43 (P31–43) of α-gliadin, activation of TcRα/β IELs by increasing their transcriptional,
which is able to upregulate major histocompatibility com- proliferative, and cytolytic activities [99]. The evidence that
plex (MHC) class I-related molecules (MICs) [94], to acti- low levels of both cytokines are found in potential CD
vate the mitogen-activated protein (MAP) kinase pathway patients supports their key role in tissue damage. The activa-
and induce apoptosis in intestinal epithelial cells. P31–43 tion of IELs, with increased Fas ligand expression, results in
induces cell proliferation and actin cytoskeleton rearrange- epithelial cell apoptosis and villous atrophy via interactions
ments in in vitro and ex vivo models [95, 96]. The prolifera- with Fas on intestinal epithelial cells.
tive response elicited by P31–43  in CD mucosa involves The end result of the above-described immune events is
epidermal growth factor (EGF)/IL-15 cooperation. In addi- the typical CD lesion characterized by intraepithelial lym-
tion, P31–43 enhances the expression of IL-15 in the small phocytosis, crypt hyperplasia, and villous atrophy. These
intestinal epithelium. IL-15 contributes to enhance the changes occur in a continuum: from a normal mucosa to a
expression of activating natural killer receptors (NKRs) on complete flattening of the villi in a slow progression. Marsh
IELs and impair the function of regulatory T cells in CD described in detail such progression [100] and his scoring
patients [97]. Activating NKRs are required for the acquisi- system is utilized by pathologists to classify CD duodenal
tion of cytolytic property by IELs. In addition to IL-15, damage as follows:
type-1 IFNs, innate cytokines promoted upon viral infec- Type 0 or pre-infiltrative stage (normal)
tions, are  responsible for inhibiting the regulatory T cell Type 1 or infiltrative stage (increased IELs)
response that counterbalance the proinflammatory T cells Type 2 or hyperplastic stage (type 1 + hyperplastic crypts)
[39]. The interplay between the adaptive gluten-specific T Type 3 or destructive stage (type 2 + villous atrophy of pro-
cell response and the innate immune pathways is key to gressively more severe degrees, denominated 3a-partial
determine the full activation of IELs that finally leads to vil- atrophy, 3b-subtotal atrophy, and 3c-total atrophy)
lous atrophy.

I ELs Activation and the Induction of Tissue A Mouse Model of CD


Damage
The profound remodeling of the small intestinal architecture To shed light on the reciprocal contribution of the innate and
typical of CD is characterized by intraepithelial lymphocyto- adaptive immune responses in the context of CD pathogen-
40  Celiac Disease 531

esis, a triple transgenic mouse model has been recently of extra-intestinal manifestations. In fact, CD presentation
developed [101]. It does not only express the human has substantially changed over time [103–106], both in chil-
HLA-DQ8 (I), one of the alleles that predisposes to CD dren and in adults, moving from a malabsorptive disorder
development, but reproduces also the overexpression of causing blatant GI symptoms and malnutrition (with failure
IL-15 in both the small intestinal epithelium (II) and the lam- to thrive being a common occurrence), to a more subtle con-
ina propria (II), similarly to what is observed in active CD dition causing a variety of extra-intestinal manifestations
patients. This animal model spontaneously develops villous that can occur either with or without a concomitant GI
atrophy after protracted gluten ingestion, which is restored involvement. Moreover, it is also well known that CD may
upon gluten withdrawal. Notably, when lacking one of the be entirely asymptomatic, as we have come to know from
three above-mentioned trans-genes, the intestinal damage screening unselected populations or first-degree relatives of
was prevented, thus showing that overexpression of IL-15 in known patients. It is no surprise therefore that the diagnostic
both mucosal compartments (epithelium and lamina propria) rate around the globe is quite dismal, as both the general
is required for the development of villous atrophy upon glu- public and the health-care providers may miss the oligo- or
ten exposure in a genetically predisposed host. Importantly, a-symptomatic individuals as well as those whose symptoms
this model allowed to demonstrate that the gluten-specific do not include obvious GI involvement [107].
CD4+ T cells, the HLA-DQ8 expression, the cytokine IFN-γ, Table 40.1 lists the main presentations of CD. As it can be
and the enzyme tTG have a crucial role in tissue destruction, seen, the clinical manifestations can be protean, thus making
suggesting new avenues for potential therapeutic strategies. the diagnosis not obvious in most cases. When CD has its
Lastly, the availability of an animal model in which the tissue onset in infancy and very early childhood, the GI manifesta-
damage develops and reverts in response to gluten provides a tions typically prevail and can be quite aggressive, resulting
key tool for testing newly developed drugs. in a clinical picture of malnutrition and failure to thrive,
often associated with a protein-losing enteropathy.
Subsequently, however, the onset may be more subtle, and
Clinical Presentations extra-intestinal manifestations become more common.

Atrophy of the villi of various degrees (from partial to total)


with crypt hyperplasia results from the inflammatory changes GI Manifestations
described in the previous section and is correlated with the
extent of the specific autoantibodies (see below under Among the most frequent GI manifestations of CD are
“Diagnosing Celiac Disease”). It should be noted however abdominal pain and bloating [108]. Chronic or intermitted
that the severity of the villous atrophy does not predict the diarrhea, characterized by bulky, foul-smelling, greasy stool,
long-term outcome of CD [102]. Malabsorption of nutrients is also a very common symptom in children with CD.  Its
thus ensues and with this, understandably, a number of gas- occurrence, however, is progressively becoming less fre-
trointestinal symptoms. But the clinical manifestations of quent than in the past [105]. Counterintuitively, long-­
celiac disease go well beyond the intestine, so that basically standing and occasionally severe constipation can be the
all systems and organs can be involved, leading to a variety presenting manifestation in a significant portion of both

Table 40.1  Main presenting signs, symptoms, and laboratory changes in celiac children
GI manifestations Extra-GI manifestations Laboratory findings
Abdominal pain, bloating Weight loss/failure to thrive Elevated CD-specific antibodies
Hepertransaminasemia
Diarrhea Delayed puberty Anemia (especially iron-deficient)
Vomiting Short stature Vitamin D deficiency
Constipation Dermatitis herpetiformis Osteopenia
Anorexia Aphthous ulcers/geographical tongue
Intussusception Dental enamel defects
Celiac crisis Osteopenia, osteoporosis
Arthritis, arthralgia
Alopecia areata
Headaches, migraine
Peripheral neuropathy
Idiopathic seizures
Sad mood/depression
Psychiatric disorders
Fatigue
532 S. Guandalini and V. Discepolo

pediatric and adult patients [108–110]. Constipation appears Recurrent intussusception, an uncommon but important
to be related to a well-documented delay in oro-cecal transit GI sign, was first described in 1997 [117] and is now a well-­
time [111] possibly caused in part by disturbed upper GI described occurrence in CD children, known to be more fre-
motor function [112]. Other presenting symptoms related to quent in undiagnosed CD children than in control
the GI tract are vomiting (especially in infants and toddlers), populations [118].
weight loss, or failure to thrive. Poor appetite, occasionally More rarely CD can have its onset with a dramatic, acute
severe like a true anorexia [113], can be an accompanying presentation, referred to as “celiac crisis.” This term describes
symptom, leading – particularly in cases of delayed diagno- a sudden onset episode of explosive diarrhea associated with
sis – to severe malnutrition and cachexia (Fig.  40.2). protein-losing enteropathy, hypoalbuminemia, and profound
However, it should be noted that children with CD can also electrolyte abnormalities with metabolic acidosis leading to
be overweight or obese, as well documented in the literature dehydration and lethargy, occurring most commonly before
[114–116], so that the absence of malnutrition should by no the diagnosis of CD and in young children, although docu-
means rule out the possibility of CD. mented in adults too [119].

Fig. 40.2  Typical GI presentation in young children. Left panel: a 19-month-old girl at the time of diagnosis. Right panel: the same patient
6 weeks later
40  Celiac Disease 533

 xtra-Intestinal Manifestations and Laboratory


E characterized – but by no means constantly – by villous atro-
Changes phy. Rare in children, DH affects mostly teenagers and adults
who present symmetrical, pruritic blisters followed by ero-
Extra-intestinal symptoms (Table 40.1) occur at similar rates sions, excoriations, and hyperpigmentation. The most involved
in children and in adults [120]. In children, short stature, sites are mainly the extensor surfaces of elbows and knees,
fatigue, and headache appear to be the most common. Failure followed by shoulders, buttocks, sacral region, and face.
of proper height gain can be an isolated initial presentation Itching of variable intensity, scratching, and burning sensation
of CD [120, 121] and can be found in up to 10% of children immediately preceding the development of lesions are com-
being investigated for short stature. If diagnosed with enough mon. The diagnosis of DH is made by a direct immunofluores-
time before the onset of puberty, it typically responds well to cence biopsy of unaffected skin in close proximity to an active
the GFD [120]. While some of the extra-intestinal manifesta- lesion [123]. Pathognomonic granular IgA deposits at the
tions such as weight loss, fatigue, short stature, and delayed dermo-epidermal junction are seen (2 D). Its treatment is
puberty can be attributed to nutritional deficiencies and their based on a strict GFD that is typically extremely effective.
metabolic consequences, others follow different and often Dapsone and/or other drugs can be used during the early phase
poorly understood pathways. of treatment, until the GFD becomes effective [124].
Dermatitis herpetiformis (DH) (Fig. 40.3) is considered the Oral manifestations are well described too, among them,
skin manifestations typical of celiac disease [122]: autoanti- dental enamel hypoplasia, geographical tongue, and aph-
bodies against epidermal transglutaminase (TG3), the sup- thous ulcers. Dental enamel hypoplasia has been reported
posed autoantigen of DH, are at play in this condition also with a prevalence ranging from 10% to 97% [125–129],

a b

c d

Fig. 40.3  Dermatitis herpetiformis [from Ref. [122], reprinted with immunofluorescence showing granular IgA deposits in the basal mem-
permission]. Typical scratched papules and macules on the elbows (a) brane zone between the epidermis and dermis (d)
and on the knees (b). Fresh small blisters on the elbow (c). Direct
534 S. Guandalini and V. Discepolo

more commonly in children. This defect is thought to be sec- study [148]. Seizures are often generalized tonic-clonic, but
ondary to nutritional deficiencies and immune disturbances partial and occasionally absence seizures are also reported.
during the period of enamel formation, mostly the first In some patients with CD and epilepsy refractory to antiepi-
7 years of life [130]. Other enamel defects that can be associ- leptic drugs, seizures have been controlled with a GFD [149,
ated to CD are enamel pitting, grooving, and partial or com- 150]. Hence, it is recommended that patients with epilepsy
plete loss of the enamel. Of note, dental enamel defects can without a clear etiology should be screened for CD, as an
be found in children in the absence of any other symptoms, early diagnosis and treatment might be beneficial. Relatively
as documented in a large epidemiological study in Italian common in children and especially in adolescents are also
children [131], and are therefore a useful screening tool. psychiatric issues [151] including anxiety, often with recur-
Aphthous ulcers can be present in children with CD [132], rent panic attacks, hallucinations, and depression that appear
although they are neither characteristic nor specific to CD to persist into adult life [151]. A moderately increased risk of
since they can also be associated with other medical condi- suicide in CD patients has also been reported [152].
tions such as inflammatory bowel disease and Behcet’s dis- Interestingly, there is some evidence that the GFD may help
ease. CD has been reported as more prevalent in children in alleviating depression in celiac adolescents [153].
with geographic tongue, a benign inflammatory condition of Elevation of liver transaminases is well described in pedi-
unknown etiology usually involving the dorsal surface and atric CD, either idiopathic or associated to autoimmune hep-
lateral borders of the tongue, than in the general population atitis, and is thought to be present in about one-third of all
[133]. As in many of the extra-intestinal manifestations, geo- children with CD [154], while CD is found in 12% of chil-
graphic tongue too can occur in the absence of GI dren with mild unexplained hypertransaminasemia. Of note,
involvement. GFD normalizes transaminase levels in most patients with
Low bone mineral density is found in many newly diag- CD within a few months.
nosed patients [134] and, in some cases, especially in adults, Anemia in CD children can be the end result of several
is advanced to osteoporosis and can be associated with frac- different, and sometimes combined, causes [155]; however,
tures [135]. The etiology of such bone alterations in CD is the single most common type of anemia is due to iron defi-
multifactorial, thought to be mostly secondary to the combi- ciency (IDA). IDA may be the only clinical abnormality
nation of intestinal malabsorption and chronic inflammation. identified in many patients and can be the presenting feature
Recently, higher serum OPG, telopeptide, and lower serum of CD, especially in older children and adults [156]. Anemia
pro-peptide have been found in adult CD patients with low at CD diagnosis is associated with more severe histological
bone mineral density, suggesting an increased bone turnover and serological presentation in children [156, 157]. However,
[136]. Low bone mineral density apparently responds well to even when asymptomatic, CD can lead to IDA; in a large
the GFD in CD children, and recent data suggest that adult series of patients with asymptomatic CD, IDA was indeed
patients too can improve their bone mineral density after found in almost half of the patients, with adults having a
some years on GFD [137]. higher incidence than children: 46% versus 35% [158].
Joint involvement, though not too common, has been The most common vitamin deficiency found in children
described in children and adults with CD [138], and it is sug- with CD at diagnosis is vitamin D [159, 160], a factor likely
gested that patients presenting with unexplained articular to contribute to the low bone mineral density [161]. Testing
manifestations be tested for CD [139, 140]. for its level is therefore recommended [162] at diagnosis,
Alopecia areata is a common form of hair loss in child- when instruction on age-appropriate intake of calcium and
hood, affecting about 1–2% of the population and character- vitamin D, in order to provide adequate replenishments,
ized by a sudden onset of patchy hair loss on the scalp. It is should be provided during nutritional counseling along with
considered an autoimmune condition, and it has been shown the need for exercise to promote bone health.
to be more common in children with CD [141], once again
even in the complete absence of GI manifestations. Good
response to the GFD has been observed [120]. Disease Associations
Children with CD have an increased frequency of neuro-
logical symptoms compared to controls [142]. The most Autoimmune and some chromosomal disorders have been
common is definitely headache [143–145] that appears in known for a long time to be associated with CD. In addition,
18% of children [145] and in most cases responds well to the several large-scale epidemiological investigations have
GFD [120]; but also peripheral neuropathy [142] and sei- revealed a growing number of additional associated condi-
zures [146] are well described; ataxia on the other hand is tions, although in most cases the reasons for such associa-
described almost exclusively in adult cases [147]. The preva- tions and their clinical relevance remain unclear (Table 40.2).
lence of epilepsy in CD children also appears to be higher Among the conditions with increased prevalence in CD,
than expected, as reported in a recent large epidemiologic mostly occurring however in adults, are asthma [163],
40  Celiac Disease 535

Table 40.2  Main conditions associated with celiac disease value for biopsy-confirmed CD or not [182]. In fact, many
Genetic/ diabetic children who come to be diagnosed with CD have
Autoimmune chromosomal minimal or no symptoms. Testing children with T1DM for
Diseases conditions disorders
CD is recommended by the American Diabetes Association;
Juvenile idiopathic Type 1 diabetes Selective IgA
arthritis mellitus deficiency the International Society for Pediatric and Adolescent
Asthma Thyroid disease: Down syndrome Diabetes; the British Society of Paediatric Gastroenterology,
 Hashimoto Hepatology and Nutrition; the European Society for Paediatric
thyroiditis Gastroenterology, Hepatology and Nutrition (ESPGHAN) in
 Graves-­
Basedow
their 2012 as well as 2020 guidelines; and the North American
disease Society for Pediatric Gastroenterology, Hepatology and
Eosinophilic esophagitis Addison disease Turner syndrome Nutrition [1, 183–187]. As patients with T1DM can however
Gastroesophageal reflux Psoriasis Williams-Beuren have mildly elevated titters of TTG-IgA independently of
syndrome CD, it is recommended to proceed with the confirmatory
Pancreatitis Systemic lupus
biopsy only when their titer is >3  times the upper limit of
Adrenal insufficiency
Idiopathic dilated
normal (ULN). Clearly, once diagnosed with CD, children
cardiomyopathy with T1DM need to follow a strict GFD; however, its effects
Atrial fibrillation on diabetic control are unclear. In fact, glycemic control, both
Cataracts in children with or without malabsorption, has been found
Uveitis either improved or unaffected [181].
Renal disease Autoimmune thyroid diseases (both Hashimoto’s thyroid-
Stroke
itis and Graves-Basedow disease) are more frequent in CD
Chronic obstructive
pulmonary disease children than in controls [188]. In these patients, GFD does
not appear to prevent the progression of the autoimmune pro-
cess [189].
Addison disease, mostly in adults, has an increased preva-
chronic obstructive pulmonary disease [164], cardiac disor- lence in CD patients as well [190]. Psoriasis appears to be
ders and strokes [165–167], gastroesophageal reflux disease more prevalent in CD patients, too [191].
[168], pancreatitis [169], ocular complications [170, 171], The issue of whether the onset of autoimmune disorders
idiopathic dilated cardiomyopathy [165], renal disease [172], in CD patients is favored by the ingestion of gluten (either
adrenal insufficiency [173], and systemic lupus [174]. before or after diagnosis) remains controversial. An increased
Of note, in children, the association with eosinophilic prevalence of autoimmune disorders was found in parallel
esophagitis has been described [169] and the effect of GFD with the increasing age at diagnosis of CD [192], suggesting
in both conditions has been assessed [175]. that prolonged exposure to gluten may indeed favor the onset
of autoimmune conditions. However, as further studies on
this issue have revealed conflicting results, the issue is still
Autoimmune Conditions unclear [193].

CD shows a strong association with autoimmune disorders,


thought to be mostly due to a shared genetic component in the Genetic/Chromosomal Disorders
HLA region [2]. The best described association is with
T1DM, a condition sharing common pathogenetic mecha- Selective IgA deficiency has also been associated with
nisms with CD [176]. Indeed, CD has a prevalence of approx- CD. In fact, about 14% of IgA-deficient children are celiac
imately 10% among T1DM patients [177–179]. About [194] and about 2% of CD children are IgA-deficient [195].
two-thirds of children diagnosed with CD after T1DM onset While clinically not relevant, this association bears impor-
have elevated levels of celiac antibodies at the time of T1DM tant implications in the strategy for diagnosing CD (see
diagnosis or within the first 24  months; however, an addi- below). CD prevalence has been found to be between five-
tional 40% of patients develop CD a few years after T1DM and tenfold higher in children with Down syndrome [3, 4],
onset [180], and even adults with a long history of T1DM Turner syndrome [5], and Williams-Beuren syndrome [6]
show a progressively higher prevalence of CD [181]. Thus, it than in the general population; hence, they need to be
is recommended that T1DM children be repeatedly tested for screened for CD. Finally, given the high prevalence among
CD. Of note, it has been shown that the presence or absence first-degree relatives of CD patients [196], they must be
of GI symptoms in children with T1DM has no predictable screened too for CD even if asymptomatic.
536 S. Guandalini and V. Discepolo

Diagnosing CD is ≥10 times the ULN and the family agrees, the no-biopsy
diagnosis may be applied, provided EMA-IgA will test posi-
Given the various clinical manifestations of CD, which can tive in a second blood sample. HLA-DQ2/-DQ8 determina-
also be asymptomatic, as well as the association with other tion and symptoms are not obligatory criteria. CD diagnosis
conditions, clearly the first requisite for diagnosis is a high can be accurately established with or without duodenal biop-
degree of suspicion. Table  40.3 lists the circumstances that sies if given recommendations are followed” [186]. It is esti-
demand testing for CD regardless of the presence or absence mated that by applying this approach, about 50% of children
of symptoms. The availability of very sensitive and specific presenting with a suspected CD can be diagnosed without a
IgA autoantibodies, generated in the small intestinal mucosa biopsy. In addition, a recent study in the USA [203] esti-
and detectable in the serum, such as the tTG-IgA or anti-­ mated that using this algorithm could avoid “between 4891
endomysium IgA (EMA-IgA) [197], has given the physician a and 7738 pediatric endoscopies per year in the United
powerful screening tool. In addition, another class of antibod- States,” thus resulting in a considerable cost saving. The
ies has also been found valuable in screening and in following algorithm presented in Fig.  40.4 (from Ref. [186] summa-
up CD patients: the anti-deamidated gliadin peptide (DGP) rizes accordingly the diagnostic steps for a child/teenager
antibodies (both IgA and IgG), produced against the gliadin suspected of CD).
peptides after they have been modified by the tTG [198]. Subjects with normal serum IgA and normal tTG-IgA CD
The first step in screening for CD in subjects of any age can be excluded, given the high sensitivity of the test; how-
who are on a gluten-containing diet is the serum level of ever, the rare seronegative CD patient has been described
tTG-IgA [1, 185, 186, 199], considered to possess the high- [204], more commonly among adults [205]. Thus, in the
est degree of sensitivity, thus basically avoiding missing any presence of a strong clinical suspicion, it would be appropri-
case of CD. Total serum IgA levels however need to be added ate to still proceed with the diagnostic biopsy even with nor-
in order to ascertain that the individual can produce tTG-IgA mal CD-specific antibodies. A recent consensus statement
and identify IgA-deficient subjects. In those cases, both tTG-­ [206] recommends that – after having ruled out alternative
IgG and DGP-IgG [200, 201] can be usefully checked as causes for villous atrophy – patients with suspected CD who
markers of CD. are seronegative but have villous atrophy and are HLA-DQ2
In 2012, an ad hoc task force of ESPGHAN published or DQ8-positive should be put on a GFD and then re-assessed
revised criteria [1] and produced an evidence-based algo- endoscopically after 1–3 years on a GFD, to evaluate resolu-
rithm that allowed the physician to skip the duodenal biopsy tion of the villous atrophy that would confirm the diagnosis.
under certain circumstances, namely, in patients showing a On another hand, if the subject has positive tTG-IgA, but at
genetic asset and a history compatible with CD and very titers lower than 10  times normal and/or with a negative
elevated titers (more than ten times the ULN) of tTG-IgA, EMA, then the EGD would be necessary.
along with a positive EMA titer. In the years that followed, During the upper endoscopy it is important to obtain at
many observations were carried out in various parts of the least four biopsies from the distal duodenum, and one or two
world, including in North America [202], basically validat- from the bulb; otherwise the diagnostic yield may be jeopar-
ing this suggested approach. Then in 2020, after gathering dized by the occasional patchy duodenal lesions. In inter-
multicenter experience, ESPGHAN went further eliminating preting the pathology of the duodenal biopsies, caution must
the need to document a consistent genetic asset as well as the be exerted for findings of Marsh type 1, especially when not
presence of CD-compatible symptoms, stating “If TGA-IgA supported by positive serology. In fact, such increase in IELs
(“lymphocytic duodenosis”) has been found to be due to CD
Table 40.3  Subjects to be screened for CD in no more than 16–39% of cases [207, 208]. Thus, addi-
All those presenting the conditions listed in Tables 40.1 and 40.2 tional conditions (Table  40.4) must be carefully ruled out
First-degree relatives of celiac patients before concluding for CD. As for those with a Marsh type 0,
Autoimmune conditions the call is even more delicate. In fact, patients could be
 Type 1 diabetes defined as “potential CD” if tTG-IgA and EMA titers are
 Autoimmune thyroiditis
 Autoimmune hepatitis positive; while in case of low titers of tTG-IgA and a nega-
 Addison’s disease tive EMA, the possibility of a false-positive tTG-IgA must
 Hashimoto Thyroiditis be considered. This occurrence is especially common, for
 Graves-Basedow disease unclear reasons, in children with T1DM, where tTG-IgA has
Genetic/chromosomal disorders
 Selective IgA deficiency
also been found to spontaneously normalize [209–211].
 Down syndrome Asymptomatic patients with mild elevation of tTG-IgA but a
 Turner syndrome normal biopsy should therefore remain on a gluten-­containing
 Williams-Beuren syndrome diet and be carefully monitored.
40  Celiac Disease 537

Potential Celiac Disease biopsy every 2 years. During the follow-up period, 42 (15%) of
280 children developed villous atrophy, whereas 89 (32%)
More complex is the decision about the need for a GFD for true children no longer tested positive for tTG-­IgA or EMA. The
“potential” CD children and adolescents. A large prospective cumulative incidence of progression to villous atrophy was
study conducted in Italy [212] with up to 12 years of follow-up 43% at 12 years. Of interest, the factors most strongly associ-
of 280 of these children investigated the possible risk factors ated with development of villous atrophy were numbers of γδ
associated with the development of villous atrophy. The chil- IELs followed by age (younger children having less chances of
dren underwent serologic tests twice a year and a small bowel developing full-blown CD) and HLA-DQ2 homozygosity.

a
CD serology positive Clinical suspicion of CD CD risk group INITIAL STAGE
for any reason1

Measure serum TGA-lgA and total lgA

TGA lgA negative


TGA-lgA & total lgA SEE B

TGA-lgA positive

Refer to paediatric GI (specialist in CD)

Review the results of the initial TGA-lgA antibodies2 SPECIALIST CARE


Discuss diagnostic path ways with the family3
Discrepant
serology
TGA-lgA SEE B

TGA-lgA positive

TGA-lgA <10x ULN4


consider TGA-lgA
conc.(xULN)

TGA-lgA 10xULN
BIOPSY SEE C
Test for EMA (separate blood sample)

EMA-lgA negative
EMA-lgA

EMA positive

CD CONFIRMED

Fig. 40.4 Algorithm for diagnosis of celiac disease [from [186], evaluation after introduction of the diet would need prolonged
reprinted with permission]. (a) in IgA-competent subjects, (b) in IgA-­ re-exposure to gluten with a series of further investigations
deficient subjects, and (c) for biopsy 4
If TGA-IgA is only borderline positive, confirm sufficient gluten
1
Other than TGA-IgA, including point-of-care tests and DGP intake and consider re-­testing of TGA-IgA and EMA
2
Check the value also in relation to the cutoff and repeat the test if 5
Low for age or <0.2 g/L above the age of 3 years
questionable or borderline. No need to retest if done with validated 6
For example, dermatitis herpetiformis, in which serology is frequently
assay with calibration curve. Test with conventional TGA-IgA test if negative
positive POCT and TGA has not been measured quantitatively 7
The cutoff for normal numbers of IEL is >25 cells/100 enterocytes.
3
Convey the message that the diagnosis of celiac disease with or with- The action done at the initial stage in primary care (light blue) is sepa-
out biopsy confirms the need for a lifelong gluten-free diet and that re- rated from specialist care (dark blue)
538 S. Guandalini and V. Discepolo

INITAL STAGE
b TGA lgA neagative

Review initial total lgA

Total lgA low5


Total lgA

Total lgA normal

Consider risk of false negative serology:


• low or short duration of gluten intake
• Immunosuppressive medication
• Extraintestinal manifestations5
Risk for false
seronegativity
Risk for false
Consult paediatric GI (specialist in CD)
seronegativity

No risk

No CD

Risk for false negative serology? Total lgA low SPECIALIST CARE

Consider HLA determination Perform lgG tests (TGA, EMA, DGP)

lgG tests positive


lgG
HLA-DQ2/DQ8 BIOPSY-SEE C
TGA, EMA or DGP

Neither DQ2 nor DQ8 HLA DQ2/8 positive lgG tests negative

Cd unlikely, consider Follow and retest Consider risk of false negative serology
other diagnosis on a normal gluten
intake, consider
biopsy Risk for false
seronegativity
Risk possible

No risk

No CD

c
SPECIALIST CARE
Esophagogastroduodenoscopy

4 biopsies from distal duodenum and


1 from bulb

Marsh 0-1
Revise quality & orientation of the biopsy
Histopathology 7 Consult experienced pathologist

Revise
Marsh 2-3
histopathology

CD confirmed
Marsh 0-1

Marsh 0 (normal) or Marsh 1 (only increased IEL)


confirmed and positive serology. Options
• Consider consultation with CD expert center
• Consider false positive TGA result and test for
EMA (if positive = potential CD)
• Consider additional tests (HLA, TGA deposits, etc.)
• Follow and retest on a normal gluten intake
• Consider relevance of symptoms

Fig. 40.4 (continued)
40  Celiac Disease 539

Table 40.4  Conditions causing lymphocytic duodenosis (Marsh type Treatment


1 changes)
Celiac disease CD is a lifelong condition and a strict GFD is currently the
H. pylori gastritis only available effective treatment. The dietary restriction
Small-bowel bacterial overgrowth
must include wheat, rye, barley, and their derivatives such as
NSAID and other drugs
Immune dysregulation triticale, spelt, and kamut. Other cereals such as rice, corn,
Crohn’s disease and buckwheat are perfectly safe for CD patients and can be
Food protein-induced enteropathy used as wheat substitutes.
Infections How strict should the diet be? While the exact amount of
NSAID (nonsteroidal anti-inflammatory drugs) gluten that can be tolerated daily by CD patients is unknown
and is likely to be subject to a wide interindividual variabil-
ity, there is evidence that a daily intake of less than 10 mg is
Thus, in practical terms the decision on whether to put a safe for all patients, while the majority of them would react
potential CD patient on a GFD must be taken with great care, at levels equal or higher than 100  mg per day [216, 217].
on an individual basis, after having properly informed the This very strict diet proves to be very hard to follow, and in
parents and obtained their full agreement. It goes without fact there are numerous reports in the literature showing that
saying that if a potential celiac is left on gluten, he/she must many celiac patients, especially in the adult population,
be periodically followed. experience an involuntary intake of gluten exceeding the safe
amounts [218, 219]. A source of concern for risk of contami-
nation with gluten in children is their school activities. A
Nonresponsive Celiac Disease recent test on surfaces that could be contaminated with glu-
ten revealed that there is indeed a potential for gluten expo-
While the GFD appears to be very effective in children, a sure at school that varied among different materials (higher
minute portion of patients would have persistent elevated for paper mache and baking products) [220]. Inadequate
serology and/or symptoms, configuring a condition of adherence to the GFD has been recently assessed in a sys-
“nonresponsive CD.” In these circumstances, a careful tematic review gathering almost 8000 children [221] that
analysis of all potential causes is needed, keeping in mind showed rates of adherence ranging from 23% to 98%. As
that by far the commonest is ongoing ingestion of gluten expected, adolescents were more at risk of nonadherence,
that can be detected and resolved by the use of a rigorously while children whose parents had good knowledge about CD
supervised and strict GFD [213]. Among the less frequent adhered more strictly. Nonadherence is associated with
causes of nonresponsive CD, recently a superimposed patient growth, symptoms, and quality of life. To explain at
cow’s milk protein allergic enteropathy has also been least in part such lack of adherence, one must note that chil-
described [214]. The term “refractory CD” (RCD) instead dren (especially older ones and teenagers) may not present
refers to the persistence of intestinal villous atrophy (with any symptom after inadvertent gluten ingestion or sporadic
or without persistent positive serology) for more than intentional consumption of gluten-containing products. On
1–2  years despite a strict GFD in patients with CD.  This the other end of the spectrum, there is some evidence that
condition occurs rarely and its diagnosis remains difficult. using “hypervigilance” in adhering to the diet may be detri-
RCD has been subdivided into two subgroups according to mental, especially in teenagers, on quality of life [222].
the normal (RCDI) or abnormal (RCDII) IELs phenotype, Additionally, the GFD often results in choice of manufac-
the latter considered as a low-­grade intraepithelial lym- tured GF products that are higher in sugar and fats and lack-
phoma with a poor prognosis [215]. Despite some debate, ing in fiber and micronutrients, so that children on GFD
there is scarce evidence for the existence of this condition experience unbalanced nutrition, from excessive caloric
in children. Hence, before concluding that a child presents intake [223] to micronutrient deficiency [223–225].
true RCD, it is strongly recommended investigating prop- Furthermore, in a study on a large number of adults, it was
erly other causes. As mentioned, in addition to ongoing found that those following a GFD had significantly higher
inadvertent ingestion of gluten, they would include condi- urine levels of total arsenic and blood levels of mercury, lead,
tions such as Crohn’s disease, autoimmune enteropathy, and cadmium, possibly as a result of larger intake of fish and
small bowel bacterial overgrowth, cow’s milk protein rice [226]. Recently, assessing the dietary preferences of a
allergy, and pancreatic insufficiency. cohort of children with CD, we find that processed food
540 S. Guandalini and V. Discepolo

items have become a staple of the GFD, and in many cases, this method, studies in adults found that gluten ingestion
these products were consumed to the exclusion of healthy occurs frequently despite efforts to follow a strict GFD
alternatives [227]. Variable effects on body mass index [240, 241]. While this is a useful tool in verifying if
(BMI) have been observed after beginning the GFD, likely recently developed symptoms can be attributed to a sus-
the result of different approaches to feeding habits in various pected, inadvertent ingestion of gluten, it must be kept in
sociocultural environments [228–230]. mind that it cannot be relied upon to verify prior, long-
Safety of consuming oats (a non-gluten-containing cereal) standing compliance.
had been a matter of debate for a very long time, with reiter- In summary, following a strict GFD is not easy, may
ated proofs  – summarized in a recent meta-analysis [231] expose to nutritional deficiencies/excesses and risks, and
that non-contaminated oats are indeed perfectly safe; in addi- carries an impact on quality of life, especially in adults. In
tion, a good source of fiber and nutrients, including oats, addition, there is a subgroup of CD patients (i.e., RCD) who
adds variety and palatability to the diet. fail to respond even to a strict GFD, thus reinforcing the con-
The GFD, particularly in pediatric age, has a strong record cept that GFD could not be considered a cure for all CD
of efficacy in resolving the presenting symptoms of CD patients. As a result, in the last decade research has focused
[232]: while GI symptoms appear to be better responding on numerous alternative forms of treatment.
than extra-intestinal ones to GFD, constipation appears to Each one of these recently reviewed [242] alternatives is
have the worst rate of improvement for both children and aimed at attacking a specific point in the pathogenesis of CD
adults at only 74% and 58%, respectively [233]. Among (Fig.  40.5): from genetically modifying the gliadin-­
extra-intestinal manifestations short stature and psychiatric containing grains in order to eliminate the toxic peptides, to
disorders showed the worst rates of improvement in children enzymatically degrading the ingested gluten (endopepti-
[233]. GFD resulted in greater rates of improvements for GI dases) in the stomach before they reach the small intestine
than for extra-GI manifestations, both children and adults (latiglutenase [243] and AN-PEP [244]); from utilizing pro-
[120, 234]. Furthermore, in general, improvement in chil- biotics to affect the microbiota in symptomatic patients on
dren was more rapid than in adults. Even more important, GFD [245] to polymers that would sequester gliadin peptides
healing of the damaged duodenal mucosa has been docu- (BL-7010) preventing them from crossing the intestinal bar-
mented in the vast majority of children after 1–2  years on rier [246]; from agents like larazotide able to modulate the
GFD [235, 236]. Thus, even if up to 20% of the patients re-­ tight junctions in order to block the entry of such peptides
biopsied in large part due to persistent symptoms (hence, a [247] to inhibiting the deamidation of gliadin peptides by the
small minority of the total) may have persistent mucosal TG2 (ZED 1227); and from blocking these peptides from
damage [237], systemic re-biopsying in CD children on adhering to the HLA-DQ2/HLA-DQ8 receptors [248] to
GFD is not recommended [238]. interfering at various levels with the subsequent steps of
Assessing dietary compliance is not always an easy immune activation by CD4 T-cells and their cytokines.
task, since serology often fails to reveal slight transgres- Among the latter, recently the development of a promising
sions. Therefore, a periodical follow-up is needed, and the therapeutic vaccine (Nexvax2) has been halted as it “did not
importance of a lifelong diet should be constantly rein- provide statistically meaningful protection from gluten expo-
forced, especially to asymptomatic patients. Recently, a sure for celiac disease patients when compared with pla-
test has been developed [239] and later marketed that cebo” [249]. All of the other approaches listed above are in
allows the detection in samples of stools (more sensitive) various phases of active development [242], from preclinical
and urine (less sensitive) of even minimal amounts of glu- to phase II clinical trials, but at this time none has been
ten that had been ingested in the previous 48 h. By using FDA-approved.
40  Celiac Disease 541

Toxic or immunogenic
gluten peptides
(wheat, barley, and rye)
1
Endopeptidases
ALV003
AN-PEP
STAN-1

2
Gluten
sequestering 4
polymer Tight junction
BL-7010 modulator
(Larazotide 3
acetate/AT-1001) Probiotics
Bifidobacterium
Lumen infantis
Mucosal layer

Tight
Epithelium junction
Apoptosis IEL

Lamina Matrix degradation and mucosal remodeling


propria Matrix (crypt hyperplasia and villous atrophy)
Mucosa

metalloproteinases MMP-1, -3, -12


9
Myofibroblast Anti-IL-15
10 7
Anti IFNγ
Gluten tolerization
Anti TNFα
Deamination Deaminated Nexvax 2, Hookworm
gluten
TG2
HLA CD4
DQ2/ DC T cell
5 DQ8 11
TG2 Anti-CD3
inhibitors Anti-CD20
Plasma
8 cell
CCR9 antagonist
6 CCX282B (CCR9, α4β7)
DQ2/DQ8 blocking
peptide analogues

Antibodies to
gluten and TG2

Fig. 40.5  New nondietary therapies for celiac disease (from Ref. [242] peptides to the far more antigenically potent deamidated gliadin pep-
with permission). (1) Endopeptidases: Latiglutenase (formerly tides. (6) DQ2/DQ8 blocking peptide analogues prevent presentation of
ALV003), AN-PEP, and STAN-1 degrade gluten into nonimmunogenic gliadin from activating T cells. (7) Gluten tolerization: Nexvax2 and
particles, thereby alleviating mucosal injury. (2) Gluten-sequestering hookworm (Necator americanus) inoculation aim to downregulate the
polymer: BL-7010 binds to intraluminal gliadin and prevents its release immune response to gluten. (8) CCR9 antagonist: CCX282B blocks
and breakdown into immunogenic peptides. (3) Probiotics: this chemokine receptor to block lymphocyte homing. (9) Anti-IL-15 is
Bifidobacterium infantis protects epithelial cells from damage caused a monoclonal antibody that may prevent immune-mediated tissue
by gliadin by downregulating the proinflammatory immune response. destruction. (10) Anti IFN-γ may prevent inflammation. (11) Anti-CD3
(4) Tight junction modulator: Larazotide acetate/AT-1001 works as a antibodies suppress gluten-activated T cells; anti-CD20 antibodies sup-
tight junction modulator to prevent gliadin-induced epithelial permea- press B cells
bility. (5) TG2 inhibitor: Blocks the transformation of native gliadin

3. Liu E, Wolter-Warmerdam K, Marmolejo J, Daniels D, Prince G,


References Hickey F.  Routine screening for celiac disease in children with
down syndrome improves case finding. J Pediatr Gastroenterol
Nutr. 2020;71(2):252–6.
1. Husby S, Koletzko S, Korponay-Szabo IR, Mearin ML, Phillips A, 4. Ostermaier KK, Weaver AL, Myers SM, Stoeckel RE, Katusic
Shamir R, et al. European Society for Pediatric Gastroenterology, SK, Voigt RG. Incidence of celiac disease in down syndrome: a
Hepatology, and Nutrition guidelines for the diagnosis of coeliac longitudinal, population-based birth cohort study. Clin Pediatr
disease. J Pediatr Gastroenterol Nutr. 2012;54(1):136–60. (Phila). 2020;59(12):1086–91.
2. Kahaly GJ, Frommer L, Schuppan D.  Celiac disease and 5. Marild K, Stordal K, Hagman A, Ludvigsson JF.  Turner syn-
endocrine autoimmunity  – the genetic link. Autoimmun Rev. drome and celiac disease: a case-control study. Pediatrics.
2018;17(12):1169–75. 2016;137(2):e20152232.
542 S. Guandalini and V. Discepolo

6. Pangallo E, Parma B, Mariani M, Cianci P, De Paoli A, Maitz S, 25. Dydensborg Sander S, Hansen AV, Stordal K, Andersen AN,
et al. Williams-Beuren syndrome and celiac disease: a real asso- Murray JA, Husby S.  Mode of delivery is not associated with
ciation? Eur J Med Genet. 2020;63(9):103999. celiac disease. Clin Epidemiol. 2018;10:323–32.
7. Singh P, Arora A, Strand TA, Leffler DA, Catassi C, Green PH, 26. Marild K, Stephansson O, Montgomery S, Murray JA, Ludvigsson
et al. Global prevalence of celiac disease: systematic review and JF. Pregnancy outcome and risk of celiac disease in offspring: a
meta-analysis. Clin Gastroenterol Hepatol. 2018;16(6):823–36. e2 nationwide case-control study. Gastroenterology. 2012;142(1):39–
8. Makharia GK, Catassi C.  Celiac disease in Asia. Gastroenterol 45. e3
Clin N Am. 2019;48(1):101–13. 27. Namatovu F, Lindkvist M, Olsson C, Ivarsson A, Sandstrom
9. Mustalahti K, Catassi C, Reunanen A, Fabiani E, Heier M, O. Season and region of birth as risk factors for coeliac disease a
McMillan S, et  al. The prevalence of celiac disease in Europe: key to the aetiology? Arch Dis Child. 2016;101(12):1114–8.
results of a centralized, international mass screening project. Ann 28. Koletzko S, Lee HS, Beyerlein A, Aronsson CA, Hummel M,
Med. 2010;42(8):587–95. Liu E, et  al. Caesarean section on the risk of celiac disease in
10. Rubio-Tapia A, Ludvigsson JF, Brantner TL, Murray JA, Everhart the offspring: the teddy study. J Pediatr Gastroenterol Nutr.
JE.  The prevalence of celiac disease in the United States. Am J 2018;66(3):417–24.
Gastroenterol. 2012;107(10):1538–44; quiz 7, 45. 29. Dydensborg Sander S, Nybo Andersen AM, Murray JA,
11. Unalp-Arida A, Ruhl CE, Choung RS, Brantner TL, Murray Karlstad O, Husby S, Stordal K.  Association between antibiot-
JA.  Lower prevalence of celiac disease and gluten-related dis- ics in the first year of life and celiac disease. Gastroenterology.
orders in persons living in southern vs northern latitudes of the 2019;156(8):2217–29.
United States. Gastroenterology. 2017;152(8):1922–32. e2 30. Jiang HY, Zhang X, Zhou YY, Jiang CM, Shi YD. Infection, anti-
12. Dixit R, Lebwohl B, Ludvigsson JF, Lewis SK, Rizkalla-Reilly biotic exposure, and risk of celiac disease: a systematic review and
N, Green PH. Celiac disease is diagnosed less frequently in young meta-analysis. J Gastroenterol Hepatol. 2020;35(4):557–66.
adult males. Dig Dis Sci. 2014;59(7):1509–12. 31. Kolodziej M, Patro-Golab B, Gieruszczak-Bialek D, Skorka A,
13. Pinkhasov RM, Wong J, Kashanian J, Lee M, Samadi DB, Piescik-Lech M, Baron R, et  al. Association between early life
Pinkhasov MM, et al. Are men shortchanged on health? Perspective (prenatal and postnatal) antibiotic administration and coeliac
on health care utilization and health risk behavior in men and disease: a systematic review. Arch Dis Child. 2019;104(11):
women in the United States. Int J Clin Pract. 2010;64(4):475–87. 1083–9.
14. Catassi C, Kryszak D, Bhatti B, Sturgeon C, Helzlsouer K, Clipp 32. Marild K, Kahrs CR, Tapia G, Stene LC, Stordal K.  Maternal
SL, et al. Natural history of celiac disease autoimmunity in a USA infections, antibiotics, and paracetamol in pregnancy and off-
cohort followed since 1974. Ann Med. 2010;42(7):530–8. spring celiac disease: a cohort study. J Pediatr Gastroenterol Nutr.
15. Dydensborg S, Toftedal P, Biaggi M, Lillevang ST, Hansen DG, 2017;64(5):730–6.
Husby S.  Increasing prevalence of coeliac disease in Denmark: 33. Kemppainen KM, Vehik K, Lynch KF, Larsson HE, Canepa RJ,
a linkage study combining national registries. Acta Paediatr. Simell V, et al. Association between early-life antibiotic use and
2012;101(2):179–84. the risk of islet or celiac disease autoimmunity. JAMA Pediatr.
16. Lohi S, Mustalahti K, Kaukinen K, Laurila K, Collin P, Rissanen 2017;171(12):1217–25.
H, et  al. Increasing prevalence of coeliac disease over time. 34. Kemppainen KM, Lynch KF, Liu E, Lonnrot M, Simell V, Briese
Aliment Pharmacol Ther. 2007;26(9):1217–25. T, et al. Factors that increase risk of celiac disease autoimmunity
17. Ludvigsson JF, Rubio-Tapia A, van Dyke CT, Melton LJ 3rd, after a gastrointestinal infection in early life. Clin Gastroenterol
Zinsmeister AR, Lahr BD, et  al. Increasing incidence of celiac Hepatol. 2017;15(5):694–702. e5
disease in a North American population. Am J Gastroenterol. 35. Myleus A, Hernell O, Gothefors L, Hammarstrom ML, Persson
2013;108(5):818–24. LA, Stenlund H, et  al. Early infections are associated with
18. Vilppula A, Kaukinen K, Luostarinen L, Krekela I, Patrikainen increased risk for celiac disease: an incident case-referent study.
H, Valve R, et  al. Increasing prevalence and high incidence of BMC Pediatr. 2012;12:194.
celiac disease in elderly people: a population-based study. BMC 36. Auricchio R, Cielo D, de Falco R, Galatola M, Bruno V,
Gastroenterol. 2009;9:49. Malamisura B, et al. Respiratory infections and the risk of celiac
19. Walker MM, Murray JA, Ronkainen J, Aro P, Storskrubb T, disease. Pediatrics. 2017;140(4):e20164102.
D’Amato M, et  al. Detection of celiac disease and lympho- 37. Karhus LL, Gunnes N, Stordal K, Bakken IJ, Tapia G, Stene LC,
cytic enteropathy by parallel serology and histopathology in a et al. Influenza and risk of later celiac disease: a cohort study of
population-­based study. Gastroenterology. 2010;139(1):112–9. 2.6 million people. Scand J Gastroenterol. 2018;53(1):15–23.
20. Ludvigsson JF, Murray JA.  Epidemiology of celiac disease. 38. Kahrs CR, Chuda K, Tapia G, Stene LC, Marild K, Rasmussen T,
Gastroenterol Clin N Am. 2019;48(1):1–18. et al. Enterovirus as trigger of coeliac disease: nested case-control
21. Ribeiro M, Rodriguez-Quijano M, Nunes FM, Carrillo JM, study within prospective birth cohort. BMJ. 2019;364:l231.
Branlard G, Igrejas G. New insights into wheat toxicity: breeding 39. Bouziat R, Hinterleitner R, Brown JJ, Stencel-Baerenwald JE,
did not seem to contribute to a prevalence of potential celiac dis- Ikizler M, Mayassi T, et  al. Reovirus infection triggers inflam-
ease’s immunostimulatory epitopes. Food Chem. 2016;213:8–18. matory responses to dietary antigens and development of celiac
22. van den Broeck HC, de Jong HC, Salentijn EM, Dekking L, Bosch disease. Science. 2017;356(6333):44–50.
D, Hamer RJ, et al. Presence of celiac disease epitopes in mod- 40. Myleus A, Stenlund H, Hernell O, Gothefors L, Hammarstrom
ern and old hexaploid wheat varieties: wheat breeding may have ML, Persson LA, et al. Early vaccinations are not risk factors for
contributed to increased prevalence of celiac disease. Theor Appl celiac disease. Pediatrics. 2012;130(1):e63–70.
Genet. 2010;121(8):1527–39. 41. Pinto-Sanchez MI, Verdu EF, Liu E, Bercik P, Green PH,
23. Kasarda DD. Can an increase in celiac disease be attributed to an Murray JA, et  al. Gluten introduction to infant feeding and risk
increase in the gluten content of wheat as a consequence of wheat of celiac disease: systematic review and meta-analysis. J Pediatr.
breeding? J Agric Food Chem. 2013;61(6):1155–9. 2016;168:132–43. e3
24. Decker E, Hornef M, Stockinger S. Cesarean delivery is associ- 42. Silano M, Agostoni C, Sanz Y, Guandalini S. Infant feeding and
ated with celiac disease but not inflammatory bowel disease in risk of developing celiac disease: a systematic review. BMJ Open.
children. Gut Microbes. 2011;2(2):91–8. 2016;6(1):e009163.
40  Celiac Disease 543

43. Lionetti E, Castellaneta S, Francavilla R, Pulvirenti A, Tonutti E, 60. Zubillaga P, Vidales MC, Zubillaga I, Ormaechea V, Garcia-Urkia
Amarri S, et al. Introduction of gluten, HLA status, and the risk of N, Vitoria JC. HLA-DQA1 and HLA-DQB1 genetic markers and
celiac disease in children. N Engl J Med. 2014;371(14):1295–303. clinical presentation in celiac disease. J Pediatr Gastroenterol
44. Vriezinga SL, Auricchio R, Bravi E, Castillejo G, Chmielewska Nutr. 2002;34(5):548–54.
A, Crespo Escobar P, et  al. Randomized feeding interven- 61. Al-Toma A, Goerres MS, Meijer JW, Pena AS, Crusius JB,
tion in infants at high risk for celiac disease. N Engl J Med. Mulder CJ. Human leukocyte antigen-DQ2 homozygosity and the
2014;371(14):1304–15. development of refractory celiac disease and enteropathy-asso-
45. Andren Aronsson C, Lee HS, Koletzko S, Uusitalo U, Yang ciated T-cell lymphoma. Clin Gastroenterol Hepatol. 2006;4(3):
J, Virtanen SM, et  al. Effects of gluten intake on risk of celiac 315–9.
disease: a case-control study on a Swedish birth cohort. Clin 62. Vader W, Stepniak D, Kooy Y, Mearin L, Thompson A, van Rood
Gastroenterol Hepatol. 2016;14(3):403–9. e3 JJ, et  al. The HLA-DQ2 gene dose effect in celiac disease is
46. Meijer CR, Discepolo V, Troncone R, Mearin ML.  Does infant directly related to the magnitude and breadth of gluten-specific T
feeding modulate the manifestation of celiac disease and type 1 cell responses. Proc Natl Acad Sci U S A. 2003;100(21):12390–5.
diabetes? Curr Opin Clin Nutr Metab Care. 2017;20(3):222–6. 63. Louka AS, Moodie SJ, Karell K, Bolognesi E, Ascher H, Greco
47. Hyytinen M, Savilahti E, Virtanen SM, Harkonen T, Ilonen J, L, et  al. A collaborative European search for non-DQA1*05-­
Luopajarvi K, et al. Avoidance of cow’s milk-based formula for DQB1*02 celiac disease loci on HLA-DR3 haplotypes: analy-
at-risk infants does not reduce development of celiac disease: a sis of transmission from homozygous parents. Hum Immunol.
randomized controlled trial. Gastroenterology. 2017;153(4):961– 2003;64(3):350–8.
70. e3 64. Pietzak MM, Schofield TC, McGinniss MJ, Nakamura
48. Szajewska H, Shamir R, Mearin L, Ribes-Koninckx C, Catassi RM.  Stratifying risk for celiac disease in a large at-risk United
C, Domellof M, et  al. Gluten introduction and the risk of coe- States population by using HLA alleles. Clin Gastroenterol
liac disease: a position paper by the European Society for Hepatol. 2009;7(9):966–71.
Pediatric Gastroenterology, Hepatology, and Nutrition. J Pediatr 65. Karell K, Louka AS, Moodie SJ, Ascher H, Clot F, Greco L, et al.
Gastroenterol Nutr. 2016;62(3):507–13. HLA types in celiac disease patients not carrying the DQA1*05-­
49. Logan K, Perkin MR, Marrs T, Radulovic S, Craven J, Flohr C, DQB1*02 (DQ2) heterodimer: results from the European Genetics
et  al. Early gluten introduction and celiac disease in the EAT Cluster on Celiac Disease. Hum Immunol. 2003;64(4):469–77.
study: a prespecified analysis of the EAT randomized clinical trial. 66. Trynka G, Hunt KA, Bockett NA, Romanos J, Mistry V, Szperl A,
JAMA Pediatr. 2020;174(11):1041–7. et al. Dense genotyping identifies and localizes multiple common
50. Lund-Blix NA, Tapia G, Marild K, Brantsaeter AL, Eggesbo M, and rare variant association signals in celiac disease. Nat Genet.
Mandal S, et al. Maternal fibre and gluten intake during pregnancy 2011;43(12):1193–201.
and risk of childhood celiac disease: the MoBa study. Sci Rep. 67. Ricano-Ponce I, Gutierrez-Achury J, Costa AF, Deelen P,
2020;10(1):16439. Kurilshikov A, Zorro MM, et  al. Immunochip meta-analysis
51. Barroso M, Beth SA, Voortman T, Jaddoe VWV, van Zelm MC, in European and Argentinian populations identifies two novel
Moll HA, et al. Dietary patterns after the weaning and lactation genetic loci associated with celiac disease. Eur J Hum Genet.
period are associated with celiac disease autoimmunity in chil- 2020;28(3):313–23.
dren. Gastroenterology. 2018;154(8):2087–96. e7 68. Zhernakova A, Withoff S, Wijmenga C. Clinical implications of
52. Valitutti F, Cucchiara S, Fasano A. Celiac disease and the microbi- shared genetics and pathogenesis in autoimmune diseases. Nat
ome. Nutrients. 2019;11(10):2403. Rev Endocrinol. 2013;9(11):646–59.
53. Ploski R, Ek J, Thorsby E, Sollid LM.  On the HLA-DQ(alpha 69. Abadie V, Sollid LM, Barreiro LB, Jabri B. Integration of genetic
1*0501, beta 1*0201)-associated susceptibility in celiac disease: and immunological insights into a model of celiac disease patho-
a possible gene dosage effect of DQB1*0201. Tissue Antigens. genesis. Annu Rev Immunol. 2011;29:493–525.
1993;41(4):173–7. 70. Stene LC, Honeyman MC, Hoffenberg EJ, Haas JE, Sokol RJ,
54. van Belzen MJ, Koeleman BP, Crusius JB, Meijer JW, Bardoel Emery L, et al. Rotavirus infection frequency and risk of celiac
AF, Pearson PL, et  al. Defining the contribution of the HLA disease autoimmunity in early childhood: a longitudinal study.
region to cis DQ2-positive coeliac disease patients. Genes Immun. Am J Gastroenterol. 2006;101(10):2333–40.
2004;5(3):215–20. 71. Atkinson MA, Chervonsky A. Does the gut microbiota have a role
55. Molberg O, Kett K, Scott H, Thorsby E, Sollid LM, Lundin in type 1 diabetes? Early evidence from humans and animal mod-
KE. Gliadin specific, HLA DQ2-restricted T cells are commonly els of the disease. Diabetologia. 2012;55(11):2868–77.
found in small intestinal biopsies from coeliac disease patients, 72. Mathis D, Benoist C.  Microbiota and autoimmune disease: the
but not from controls. Scand J Immunol. 1997;46(3):103–9. hosted self. Cell Host Microbe. 2011;10(4):297–301.
56. Molberg O, McAdam SN, Korner R, Quarsten H, Kristiansen C, 73. Dolcino M, Zanoni G, Bason C, Tinazzi E, Boccola E, Valletta
Madsen L, et al. Tissue transglutaminase selectively modifies glia- E, et al. A subset of anti-rotavirus antibodies directed against the
din peptides that are recognized by gut-derived T cells in celiac viral protein VP7 predicts the onset of celiac disease and induces
disease. Nat Med. 1998;4(6):713–7. typical features of the disease in the intestinal epithelial cell line
57. Mearin ML, Biemond I, Pena AS, Polanco I, Vazquez C, Schreuder T84. Immunol Res. 2013;56(2–3):465–76.
GT, et al. HLA-DR phenotypes in Spanish coeliac children: their 74. Marild K, Kahrs CR, Tapia G, Stene LC, Stordal K.  Infections
contribution to the understanding of the genetics of the disease. and risk of celiac disease in childhood: a prospective nationwide
Gut. 1983;24(6):532–7. cohort study. Am J Gastroenterol. 2015;110(10):1475–84.
58. Megiorni F, Mora B, Bonamico M, Barbato M, Nenna R, Maiella 75. Tjernberg AR, Ludvigsson JF.  Children with celiac disease are
G, et  al. HLA-DQ and risk gradient for celiac disease. Hum more likely to have attended hospital for prior respiratory syncy-
Immunol. 2009;70(1):55–9. tial virus infection. Dig Dis Sci. 2014;59(7):1502–8.
59. Congia M, Cucca F, Frau F, Lampis R, Melis L, Clemente MG, 76. Forsberg G, Fahlgren A, Horstedt P, Hammarstrom S, Hernell
et al. A gene dosage effect of the DQA1*0501/DQB1*0201 allelic O, Hammarstrom ML.  Presence of bacteria and innate immu-
combination influences the clinical heterogeneity of celiac dis- nity of intestinal epithelium in childhood celiac disease. Am J
ease. Hum Immunol. 1994;40(2):138–42. Gastroenterol. 2004;99(5):894–904.
544 S. Guandalini and V. Discepolo

77. Sanz Y, Sanchez E, Marzotto M, Calabuig M, Torriani S, 96. Nanayakkara M, Lania G, Maglio M, Discepolo V, Sarno M, Gaito
Dellaglio F.  Differences in faecal bacterial communities in coe- A, et al. An undigested gliadin peptide activates innate immunity
liac and healthy children as detected by PCR and denaturing and proliferative signaling in enterocytes: the role in celiac dis-
gradient gel electrophoresis. FEMS Immunol Med Microbiol. ease. Am J Clin Nutr. 2013;98(4):1123–35.
2007;51(3):562–8. 97. Zanzi D, Stefanile R, Santagata S, Iaffaldano L, Iaquinto G,
78. Caminero A, Verdu EF.  Celiac disease: should we care Giardullo N, et  al. IL-15 interferes with suppressive activity of
about microbes? Am J Physiol Gastrointest Liver Physiol. intestinal regulatory T cells expanded in Celiac disease. Am J
2019;317(2):G161–G70. Gastroenterol. 2011;106(7):1308–17.
79. D’Argenio V, Casaburi G, Precone V, Pagliuca C, Colicchio R, 98. Mayassi T, Ladell K, Gudjonson H, McLaren JE, Shaw DG, Tran
Sarnataro D, et al. Metagenomics reveals dysbiosis and a poten- MT, et  al. Chronic inflammation permanently reshapes tissue-­
tially pathogenic N. flavescens strain in duodenum of adult celiac resident immunity in celiac disease. Cell. 2019;176(5):967–81.
patients. Am J Gastroenterol. 2016;111(6):879–90. e19
80. Olivares M, Benitez-Paez A, de Palma G, Capilla A, Nova E, 99. Ciszewski C, Discepolo V, Pacis A, Doerr N, Tastet O, Mayassi T,
Castillejo G, et al. Increased prevalence of pathogenic bacteria in et al. Identification of a γc receptor antagonist that prevents repro-
the gut microbiota of infants at risk of developing celiac disease: gramming of human tissue-resident cytotoxic T cells by IL15 and
the PROFICEL study. Gut Microbes. 2018;9(6):551–8. IL21. Gastroenterology. 2020;158(3):625–37. e13
81. Olivares M, Neef A, Castillejo G, Palma GD, Varea V, Capilla A, 100. Marsh MN.  Gluten, major histocompatibility complex, and the
et al. The HLA-DQ2 genotype selects for early intestinal micro- small intestine. A molecular and immunobiologic approach to the
biota composition in infants at high risk of developing coeliac dis- spectrum of gluten sensitivity (‘celiac sprue’). Gastroenterology.
ease. Gut. 2015;64(3):406–17. 1992;102(1):330–54.
82. Olivares M, Walker AW, Capilla A, Benitez-Paez A, Palau F, 101. Abadie V, Kim SM, Lejeune T, Palanski BA, Ernest JD, Tastet
Parkhill J, et al. Gut microbiota trajectory in early life may predict O, et al. IL-15, gluten and HLA-DQ8 drive tissue destruction in
development of celiac disease. Microbiome. 2018;6(1):36. coeliac disease. Nature. 2020;578(7796):600–4.
83. Jabri B, Sollid LM. Tissue-mediated control of immunopathology 102. Kröger S, Kurppa K, Repo M, Huhtala H, Kaukinen K, Lindfors
in coeliac disease. Nat Rev Immunol. 2009;9(12):858–70. K, et  al. Severity of villous atrophy at diagnosis in childhood
84. Abadie V, Discepolo V, Jabri B.  Intraepithelial lymphocytes does not predict long-term outcomes in celiac disease. J Pediatr
in celiac disease immunopathology. Semin Immunopathol. Gastroenterol Nutr. 2020;71(1):71–7.
2012;34(4):551–66. 103. Almallouhi E, King KS, Patel B, Wi C, Juhn YJ, Murray JA, et al.
85. Setty M, Discepolo V, Abadie V, Kamhawi S, Mayassi T, Kent A, Increasing incidence and altered presentation in a population-­
et al. Distinct and synergistic contributions of epithelial stress and based study of pediatric celiac disease in North America. J Pediatr
adaptive immunity to functions of intraepithelial killer cells and Gastroenterol Nutr. 2017;65(4):432–7.
active celiac disease. Gastroenterology. 2015;149(3):681–91. e10 104. Krauthammer A, Guz-Mark A, Zevit N, Marderfeld L, Waisbourd-­
86. Shan L, Molberg O, Parrot I, Hausch F, Filiz F, Gray GM, et al. Zinman O, Silbermintz A, et al. Two decades of pediatric celiac
Structural basis for gluten intolerance in celiac sprue. Science. disease in a tertiary referral center: what has changed? Dig Liver
2002;297(5590):2275–9. Dis. 2020;52(4):457–61.
87. Monsuur AJ, de Bakker PI, Alizadeh BZ, Zhernakova A, Bevova 105. Popp A, Maki M.  Changing pattern of childhood celiac disease
MR, Strengman E, et  al. Myosin IXB variant increases the risk epidemiology: contributing factors. Front Pediatr. 2019;7:357.
of celiac disease and points toward a primary intestinal barrier 106. Tapsas D, Hollen E, Stenhammar L, Falth-Magnusson K.  The
defect. Nat Genet. 2005;37(12):1341–4. clinical presentation of coeliac disease in 1030 Swedish chil-
88. Schumann M, Richter JF, Wedell I, Moos V, Zimmermann-­ dren: changing features over the past four decades. Dig Liver Dis.
Kordmann M, Schneider T, et al. Mechanisms of epithelial trans- 2016;48(1):16–22.
location of the alpha(2)-gliadin-33mer in coeliac sprue. Gut. 107. Paez MA, Gramelspacher AM, Sinacore J, Winterfield L, Venu
2008;57(6):747–54. M. Delay in diagnosis of celiac disease in patients without gastro-
89. Kim CY, Quarsten H, Bergseng E, Khosla C, Sollid LM. Structural intestinal complaints. Am J Med. 2017;130(11):1318–23.
basis for HLA-DQ2-mediated presentation of gluten epitopes in 108. Khatib M, Baker RD, Ly EK, Kozielski R, Baker SS. Presenting
celiac disease. Proc Natl Acad Sci U S A. 2004;101(12):4175–9. pattern of pediatric celiac disease. J Pediatr Gastroenterol Nutr.
90. Mesin L, Sollid LM, Di Niro R. The intestinal B-cell response in 2016;62(1):60–3.
celiac disease. Front Immunol. 2012;3:313. 109. Ehsani-Ardakani MJ, Rostami Nejad M, Villanacci V, Volta U,
91. Sollid LM, Tye-Din JA, Qiao SW, Anderson RP, Gianfrani C, Manenti S, Caio G, et al. Gastrointestinal and non-­gastrointestinal
Koning F.  Update 2020: nomenclature and listing of celiac presentation in patients with celiac disease. Arch Iran Med.
disease-­relevant gluten epitopes recognized by CD4(+) T cells. 2013;16(2):78–82.
Immunogenetics. 2020;72(1–2):85–8. 110. Akman S, Sahaloglu O, Dalkan C, Bahceciler NN, Arikan C. Is
92. Sperandeo MP, Tosco A, Izzo V, Tucci F, Troncone R, Auricchio celiac disease misdiagnosed in children with functional constipa-
R, et al. Potential celiac patients: a model of celiac disease patho- tion? Turk J Gastroenterol. 2018;29(2):210–4.
genesis. PLoS One. 2011;6(7):e21281. 111. Benini F, Mora A, Turini D, Bertolazzi S, Lanzarotto F, Ricci
93. Maiuri L, Ciacci C, Ricciardelli I, Vacca L, Raia V, Auricchio C, et  al. Slow gallbladder emptying reverts to normal but small
S, et  al. Association between innate response to gliadin and intestinal transit of a physiological meal remains slow in celiac
activation of pathogenic T cells in coeliac disease. Lancet. patients during gluten-free diet. Neurogastroenterol Motil.
2003;362(9377):30–7. 2012;24(2):100–7. e79–80
94. Hue S, Mention JJ, Monteiro RC, Zhang S, Cellier C, Schmitz J, 112. Cucchiara S, Bassotti G, Castellucci G, Minella R, Betti C,
et al. A direct role for NKG2D/MICA interaction in villous atro- Fusaro C, et  al. Upper gastrointestinal motor abnormalities in
phy during celiac disease. Immunity. 2004;21(3):367–77. children with active celiac disease. J Pediatr Gastroenterol Nutr.
95. Nanayakkara M, Kosova R, Lania G, Sarno M, Gaito A, Galatola 1995;21(4):435–42.
M, et al. A celiac cellular phenotype, with altered LPP sub-cellular 113. Marild K, Stordal K, Bulik CM, Rewers M, Ekbom A, Liu E,
distribution, is inducible in controls by the toxic gliadin peptide et  al. Celiac disease and anorexia nervosa: a nationwide study.
P31-43. PLoS One. 2013;8(11):e79763. Pediatrics. 2017;139(5):26.
40  Celiac Disease 545

114. Calcaterra V, Regalbuto C, Manuelli M, Klersy C, Pelizzo G, 134. Webster J, Vajravelu ME, Choi C, Zemel B, Verma R. Prevalence
Albertini R, et  al. Screening for celiac disease among children of and risk factors for low bone mineral density in children
with overweight and obesity: toward exploring celiac iceberg. with celiac disease. Clin Gastroenterol Hepatol. 2019;17(8):
J Pediatr Endocrinol Metab. 2020; https://doi.org/10.1515/ 1509–14.
jpem-­2020-­0076. 135. Duerksen DR, Lix LM, Johansson H, McCloskey EV, Harvey
115. Nenna R, Mosca A, Mennini M, Papa RE, Petrarca L, Mercurio R, NC, Kanis JA, et al. Fracture risk assessment in celiac disease: a
et al. Coeliac disease screening among a large cohort of overweight/ registry-based cohort study. Osteoporos Int. 2020;32(1):93–9.
obese children. J Pediatr Gastroenterol Nutr. 2015;60(3):405–7. 136. Di Stefano M, Bergonzi M, Benedetti I, De Amici M, Torre C,
116. Diamanti A, Capriati T, Basso MS, Panetta F, Di Ciommo Laurora Brondino N, et al. Alterations of inflammatory and matrix produc-
VM, Bellucci F, et al. Celiac disease and overweight in children: tion indices in celiac disease with low bone mass on long-term
an update. Nutrients. 2014;6(1):207–20. gluten-free diet. J Clin Gastroenterol. 2019;53(6):e221–e6.
117. Germann R, Kuch M, Prinz K, Ebbing A, Schindera F. Celiac dis- 137. Haere P, Hoie O, Lundin KEA, Haugeberg G. No major reduction
ease: an uncommon cause of recurrent intussusception. J Pediatr in bone mineral density after long-term treatment of patients with
Gastroenterol Nutr. 1997;25(4):415–6. Celiac disease. Eur J Intern Med. 2019;68:23–9.
118. Borkar VV, Poddar U, Thakral A, Agarwal J, Srivastava A, Yachha 138. Dos Santos S, Liote F.  Osteoarticular manifestations of celiac
SK, et al. Intussusception in celiac disease: is it a common feature disease and non-celiac gluten hypersensitivity. Joint Bone Spine.
in children? J Gastroenterol Hepatol. 2018;33(2):380–4. 2017;84(3):263–6.
119. Balaban DV, Dima A, Jurcut C, Popp A, Jinga M. Celiac crisis, a 139. Ghozzi M, Sakly W, Mankai A, Bouajina E, Bahri F, Nouira R,
rare occurrence in adult celiac disease: a systematic review. World et  al. Screening for celiac disease, by endomysial antibodies, in
J Clin Cases. 2019;7(3):311–9. patients with unexplained articular manifestations. Rheumatol Int.
120. Jericho H, Sansotta N, Guandalini S.  Extraintestinal manifes- 2013;34(5):637–42.
tations of celiac disease: effectiveness of the gluten-free diet. J 140. Sherman Y, Karanicolas R, DiMarco B, Pan N, Adams AB,
Pediatr Gastroenterol Nutr. 2017;65(1):75–9. Barinstein LV, et  al. Unrecognized Celiac disease in chil-
121. Troncone R, Kosova R.  Short stature and catch-up growth in dren presenting for rheumatology evaluation. Pediatrics.
celiac disease. J Pediatr Gastroenterol Nutr. 2010;51:S137–8. 2015;136(1):e68–75.
122. Reunala T, Salmi TT, Hervonen K, Kaukinen K, Collin 141. Ertekin V, Tosun MS, Erdem T. Screening of celiac disease in chil-
P. Dermatitis herpetiformis: a common extraintestinal manifesta- dren with alopecia areata. Indian J Dermatol. 2014;59(3):317.
tion of coeliac disease. Nutrients. 2018;10(5):602. 142. Mearns ES, Taylor A, Thomas Craig KJ, Puglielli S, Leffler
123. Zone JJ, Meyer LJ, Petersen MJ. Deposition of granular IgA rela- DA, Sanders DS, et al. Neurological manifestations of neuropa-
tive to clinical lesions in dermatitis herpetiformis. Arch Dermatol. thy and Ataxia in celiac disease: a systematic review. Nutrients.
1996;132(8):912–8. 2019;11(2):380.
124. Bolotin D, Petronic-Rosic V.  Dermatitis herpetiformis. Part 143. Dimitrova AK, Ungaro RC, Lebwohl B, Lewis SK, Tennyson
II. Diagnosis, management, and prognosis. J Am Acad Dermatol. CA, Green MW, et  al. Prevalence of migraine in patients with
2011;64(6):1027–33. quiz 33–4 celiac disease and inflammatory bowel disease. Headache.
125. Pastore L, Carroccio A, Compilato D, Panzarella V, Serpico R, Lo 2013;53(2):344–55.
ML. Oral manifestations of celiac disease. J Clin Gastroenterol. 144. Nenna R, Petrarca L, Verdecchia P, Florio M, Pietropaoli N,
2008;42(3):224–32. Mastrogiorgio G, et al. Celiac disease in a large cohort of children
126. Wierink CD, van Diermen DE, Aartman IH, Heymans HS. Dental and adolescents with recurrent headache: a retrospective study.
enamel defects in children with coeliac disease. Int J Paediatr Dig Liver Dis. 2016;48(5):495–8.
Dent. 2007;17(3):163–8. 145. Zis P, Julian T, Hadjivassiliou M. Headache associated with coe-
127. Avsar A, Kalayci AG.  The presence and distribution of dental liac disease: a systematic review and meta-analysis. Nutrients.
enamel defects and caries in children with celiac disease. Turk J 2018;10(10):1445.
Pediatr. 2008;50(1):45–50. 146. Bashiri H, Afshari D, Babaei N, Ghadami MR. Celiac disease and
128. Acar S, Yetkiner AA, Ersin N, Oncag O, Aydogdu S, Arikan epilepsy: the effect of gluten-free diet on seizure control. Adv Clin
C.  Oral findings and salivary parameters in children with celiac Exp Med. 2016;25(4):751–4.
disease: a preliminary study. Med Princ Pract. 2012;21(2):129–33. 147. Ruggieri M, Incorpora G, Polizzi A, Parano E, Spina M, Pavone
129. Souto-Souza D, da Consolacao Soares ME, Rezende VS, de P. Low prevalence of neurologic and psychiatric manifestations in
Lacerda Dantas PC, Galvao EL, Falci SGM. Association between children with gluten sensitivity. J Pediatr. 2008;152(2):244–9.
developmental defects of enamel and celiac disease: a meta-­ 148. Canova C, Ludvigsson J, Barbiellini Amidei C, Zanier L, Zingone
analysis. Arch Oral Biol. 2018;87:180–90. F. The risk of epilepsy in children with celiac disease: a population-­
130. Cheng J, Malahias T, Brar P, Minaya MT, Green PH. The asso- based cohort study. Eur J Neurol. 2020;27(6):1089–95.
ciation between celiac disease, dental enamel defects, and aph- 149. Canales P, Mery VP, Larrondo FJ, Bravo FL, Godoy J. Epilepsy
thous ulcers in a United States cohort. J Clin Gastroenterol. and celiac disease: favorable outcome with a gluten-free diet
2010;44(3):191–4. in a patient refractory to antiepileptic drugs. Neurologist.
131. Martelossi S, Zanatta E, Del Santo E, Clarich P, Radovich P, 2006;12(6):318–21.
Ventura A. Dental enamel defects and screening for coeliac dis- 150. Pascotto A, Coppola G, Ecuba P, Liguori G, Guandalini S.

ease. Acta Paediatr Suppl. 1996;412:47–8. Epilepsy and occipital calcifications with or without celiac dis-
132. Zoumpoulakis M, Fotoulaki M, Topitsoglou V, Lazidou P, ease: report of four cases. J Epilepsy. 1994;7:130–6.
Zouloumis L, Kotsanos N. Prevalence of dental enamel defects, 151. Lebwohl B, Haggard L, Emilsson L, Soderling J, Roelstraete B,
aphthous-like ulcers and other oral manifestations in celiac chil- Butwicka A, et  al. Psychiatric disorders in patients with a diag-
dren and adolescents: a comparative study. J Clin Pediatr Dent. nosis of celiac disease during childhood from 1973 to 2016. Clin
2019;43(4):274–80. Gastroenterol Hepatol. 2020;S1542-3565(20):31127.
133. Cigic L, Galic T, Kero D, Simunic M, Medvedec Mikic I, Kalibovic 152. Ludvigsson JF, Sellgren C, Runeson B, Langstrom N, Lichtenstein
Govorko D, et al. The prevalence of celiac disease in patients with P. Increased suicide risk in coeliac disease–a Swedish nationwide
geographic tongue. J Oral Pathol Med. 2016;45(10):791–6. cohort study. Dig Liver Dis. 2011;43(8):616–22.
546 S. Guandalini and V. Discepolo

153. Simsek S, Baysoy G, Gencoglan S, Uluca U.  Effects of gluten-­ 173. Elfstrom P, Montgomery SM, Kampe O, Ekbom A, Ludvigsson
free diet on quality of life and depression in children with celiac JF.  Risk of primary adrenal insufficiency in patients with celiac
disease. J Pediatr Gastroenterol Nutr. 2015;61(3):303–6. disease. J Clin Endocrinol Metab. 2007;92(9):3595–8.
154. Vajro P, Paolella G, Maggiore G, Giordano G.  Meta-analysis: 174. Ludvigsson JF, Rubio-Tapia A, Chowdhary V, Murray JA,
pediatric celiac disease, cryptogenic hypertransaminase- Simard JF.  Increased risk of systemic lupus erythematosus in
mia, and autoimmune hepatitis. J Pediatr Gastroenterol Nutr. 29,000 patients with biopsy-verified celiac disease. J Rheumatol.
2013;56(6):663–70. 2012;39(10):1964–70.
155. Harper JW, Holleran SF, Ramakrishnan R, Bhagat G, Green 175. Patton T, Chugh A, Padhye L, DeGeeter C, Guandalini S. Pediatric
PH. Anemia in celiac disease is multifactorial in etiology. Am J celiac disease and eosinophilic esophagitis: outcome of dietary
Hematol. 2007;82(11):996–1000. therapy. J Pediatr Gastroenterol Nutr. 2019;69(2):e43–e8.
156. Rajalahti T, Repo M, Kivela L, Huhtala H, Maki M, Kaukinen K, 176. Goodwin G. Type 1 diabetes mellitus and celiac disease: distinct
et al. Anemia in pediatric celiac disease: association with clinical autoimmune disorders that share common pathogenic mecha-
and histological features and response to gluten-free diet. J Pediatr nisms. Horm Res Paediatr. 2019;92(5):285–92.
Gastroenterol Nutr. 2017;64(1):e1–6. 177. Weiss B, Pinhas-Hamiel O. Celiac disease and diabetes: when to
157. Zanini B, Caselani F, Magni A, Turini D, Ferraresi A, Lanzarotto test and treat. J Pediatr Gastroenterol Nutr. 2017;64(2):175–9.
F, et al. Celiac disease with mild enteropathy is not mild disease. 178. Nederstigt C, Uitbeijerse BS, Janssen LGM, Corssmit EPM, de
Clin Gastroenterol Hepatol. 2013;11(3):253–8. Koning EJP, Dekkers OM.  Associated auto-immune disease in
158. Bottaro G, Cataldo F, Rotolo N, Spina M, Corazza GR. The clini- type 1 diabetes patients: a systematic review and meta-analysis.
cal pattern of subclinical/silent celiac disease: an analysis on 1026 Eur J Endocrinol. 2019;180(2):135–44.
consecutive cases. Am J Gastroenterol. 1999;94(3):691–6. 179. Hagopian W, Lee HS, Liu E, Rewers M, She JX, Ziegler AG, et al.
159. Deora V, Aylward N, Sokoro A, El-Matary W. Serum vitamins and Co-occurrence of type 1 diabetes and celiac disease autoimmu-
minerals at diagnosis and follow-up in children with celiac dis- nity. Pediatrics. 2017;140(5):e20171305.
ease. J Pediatr Gastroenterol Nutr. 2017;65(2):185–9. 180. Barera G, Bonfanti R, Viscardi M, Bazzigaluppi E, Calori G,
160. Villanueva J, Maranda L, Nwosu BU. Is vitamin D deficiency a Meschi F, et al. Occurrence of celiac disease after onset of type
feature of pediatric celiac disease? J Pediatr Endocrinol Metab. 1 diabetes: a 6-year prospective longitudinal study. Pediatrics.
2012;25(5–6):607–10. 2002;109(5):833–8.
161. Mager DR, Qiao J, Turner J. Vitamin D and K status influences 181. Scaramuzza AE, Mantegazza C, Bosetti A, Zuccotti GV. Type 1
bone mineral density and bone accrual in children and adolescents diabetes and celiac disease: the effects of gluten free diet on meta-
with celiac disease. Eur J Clin Nutr. 2012;66(4):488–95. bolic control. World J Diabetes. 2013;4(4):130–4.
162. Snyder J, Butzner JD, DeFelice AR, Fasano A, Guandalini 182. Bybrant MC, Ortqvist E, Lantz S, Grahnquist L. High prevalence
S, Liu E, et  al. Evidence-informed expert recommendations of celiac disease in Swedish children and adolescents with type
for the management of celiac disease in children. Pediatrics. 1 diabetes and the relation to the Swedish epidemic of celiac
2016;138(3):e20153147. disease: a cohort study. Scand J Gastroenterol. 2014;49(1):
163. Ludvigsson JF, Hemminki K, Wahlstrom J, Almqvist C.  Celiac 52–8.
disease confers a 1.6-fold increased risk of asthma: a nation- 183. Couper JJ, Haller MJ, Ziegler AG, Knip M, Ludvigsson J, Craig
wide population-based cohort study. J Allergy Clin Immunol. ME, et al. ISPAD Clinical Practice Consensus Guidelines 2014.
2011;127(4):1071–3. Phases of type 1 diabetes in children and adolescents. Pediatr
164. Ludvigsson JF, Inghammar M, Ekberg M, Egesten A. A nation- Diabetes. 2014;15(Suppl 20):18–25.
wide cohort study of the risk of chronic obstructive pulmonary 184. Hill ID, Dirks MH, Liptak GS, Colletti RB, Fasano A, Guandalini
disease in coeliac disease. J Intern Med. 2012;271(5):481–9. S, et al. Guideline for the diagnosis and treatment of celiac disease
165. Emilsson L, Andersson B, Elfstrom P, Green PH, Ludvigsson in children: recommendations of the North American Society for
JF. Risk of idiopathic dilated cardiomyopathy in 29 000 patients Pediatric Gastroenterology, Hepatology and Nutrition. J Pediatr
with celiac disease. J Am Heart Assoc. 2012;1(3):e001594. Gastroenterol Nutr. 2005;40(1):1–19.
166. Emilsson L, Smith JG, West J, Melander O, Ludvigsson 185. Hill ID, Fasano A, Guandalini S, Hoffenberg E, Levy J, Reilly
JF. Increased risk of atrial fibrillation in patients with coeliac dis- N, et al. NASPGHAN clinical report on the diagnosis and treat-
ease: a nationwide cohort study. Eur Heart J. 2011;32(19):2430–7. ment of gluten-related disorders. J Pediatr Gastroenterol Nutr.
167. Ludvigsson JF, West J, Card T, Appelros P.  Risk of stroke in 2016;63(1):156–65.
28,000 patients with celiac disease: a nationwide cohort study in 186. Husby S, Koletzko S, Korponay-Szabo I, Kurppa K, Mearin
Sweden. J Stroke Cerebrovasc Dis. 2012;21(8):860–7. ML, Ribes-Koninckx C, et  al. European society paediatric gas-
168. Ludvigsson JF, Aro P, Walker MM, Vieth M, Agreus L, Talley NJ, troenterology, hepatology and nutrition guidelines for diagnosing
et  al. Celiac disease, eosinophilic esophagitis and gastroesopha- coeliac disease 2020. J Pediatr Gastroenterol Nutr. 2020;70(1):
geal reflux disease, an adult population-based study. Scand J 141–56.
Gastroenterol. 2013;48(7):808–14. 187. Murch S, Jenkins H, Auth M, Bremner R, Butt A, France S, et al.
169. Sadr-Azodi O, Sanders DS, Murray JA, Ludvigsson JF. Patients Joint BSPGHAN and Coeliac UK guidelines for the diagnosis
with celiac disease have an increased risk for pancreatitis. Clin and management of coeliac disease in children. Arch Dis Child.
Gastroenterol Hepatol. 2012;10(10):1136–42. e3 2013;98(10):806–11.
170. Mollazadegan K, Kugelberg M, Lindblad BE, Ludvigsson 188. Minelli R, Gaiani F, Kayali S, Di Mario F, Fornaroli F, Leandro
JF. Increased risk of cataract among 28,000 patients with celiac G, et al. Thyroid and celiac disease in pediatric age: a literature
disease. Am J Epidemiol. 2011;174(2):195–202. review. Acta Biomed. 2018;89(9-S):11–6.
171. Mollazadegan K, Kugelberg M, Tallstedt L, Ludvigsson 189. Metso S, Hyytia-Ilmonen H, Kaukinen K, Huhtala H, Jaatinen
JF. Increased risk of uveitis in coeliac disease: a nationwide cohort P, Salmi J, et al. Gluten-free diet and autoimmune thyroiditis in
study. Br J Ophthalmol. 2012;96(6):857–61. patients with celiac disease. A prospective controlled study. Scand
172. Wijarnpreecha K, Thongprayoon C, Panjawatanan P, Thamcharoen J Gastroenterol. 2012;47(1):43–8.
N, Pachariyanon P, Nakkala K, et al. Celiac disease and the risk of 190. O’Leary C, Walsh CH, Wieneke P, O’Regan P, Buckley B,
kidney diseases: a systematic review and meta-analysis. Dig Liver O’Halloran DJ, et al. Coeliac disease and autoimmune Addison’s
Dis. 2016;48(12):1418–24. disease: a clinical pitfall. QJM. 2002;95(2):79–82.
40  Celiac Disease 547

191. De Bastiani R, Gabrielli M, Lora L, Napoli L, Tosetti C, Pirrotta 208. Santolaria S, Dominguez M, Alcedo J, Abascal M, Garcia-­
E, et al. Association between coeliac disease and psoriasis: Italian Prats MD, Marigil M, et  al. Lymphocytic duodenosis: etio-
primary care multicentre study. Dermatology. 2015;230(2): logical study and clinical presentations. Gastroenterol Hepatol.
156–60. 2013;36(9):565–73.
192. Ventura A, Magazzu G, Greco L.  Duration of exposure to glu- 209. Castellaneta S, Piccinno E, Oliva M, Cristofori F, Vendemiale M,
ten and risk for autoimmune disorders in patients with celiac dis- Ortolani F, et al. High rate of spontaneous normalization of celiac
ease. SIGEP Study Group for Autoimmune Disorders in Celiac serology in a cohort of 446 children with type 1 diabetes: a pro-
Disease. Gastroenterology. 1999;117(2):297–303. spective study. Diabetes Care. 2015;38(5):760–6.
193. Elli L, Discepolo V, Bardella MT, Guandalini S.  Does gluten 210. Waisbourd-Zinman O, Hojsak I, Rosenbach Y, Mozer-Glassberg
intake influence the development of celiac disease-associated Y, Shalitin S, Phillip M, et al. Spontaneous normalization of anti-­
complications? J Clin Gastroenterol. 2014;48(1):13–20. tissue transglutaminase antibody levels is common in children
194. Lougaris V, Sorlini A, Monfredini C, Ingrasciotta G, Caravaggio with type 1 diabetes mellitus. Dig Dis Sci. 2012;57(5):1314–20.
A, Lorenzini T, et  al. Clinical and laboratory features of 184 211. Unal E, Demiral M, Baysal B, Agin M, Devecioglu EG,
Italian pediatric patients affected with selective IgA deficiency Demirbilek H, et  al. Frequency of celiac disease and spontane-
(SIgAD): a longitudinal single-center study. J Clin Immunol. ous normalization rate of celiac serology in children and adoles-
2019;39(5):470–5. cent patients with type 1 diabetes. J Clin Res Pediatr Endocrinol.
195. Cataldo F, Marino V, Bottaro G, Greco P, Ventura A.  Celiac 2020;13(1):72–9.
disease and selective immunoglobulin A deficiency. J Pediatr. 212. Auricchio R, Mandile R, Del Vecchio MR, Scapaticci S, Galatola
1997;131(2):306–8. M, Maglio M, et al. Progression of celiac disease in children with
196. Nellikkal SS, Hafed Y, Larson JJ, Murray JA, Absah I.  High antibodies against tissue transglutaminase and normal duodenal
prevalence of celiac disease among screened first-degree relatives. architecture. Gastroenterology. 2019;157(2):413–20. e3
Mayo Clin Proc. 2019;94(9):1807–13. 213. Leonard MM, Cureton P, Fasano A.  Indications and use of the
197. Giersiepen K, Lelgemann M, Stuhldreher N, Ronfani L, Husby S, gluten contamination elimination diet for patients with non-­
Koletzko S, et al. Accuracy of diagnostic antibody tests for coe- responsive celiac disease. Nutrients. 2017;9(10):1129.
liac disease in children: summary of an evidence report. J Pediatr 214. Capone K, Sansotta N, Vohra P, Jericho H, Guandalini S.  Milk
Gastroenterol Nutr. 2012;54(2):229–41. protein-induced villous atrophy and elevated serologies in four
198. Aleanzi M, Demonte AM, Esper C, Garcilazo S, Waggener children with celiac disease on a gluten-free diet. Ann Pediatr.
M.  Celiac disease: antibody recognition against native 2020;3(1):1–4.
and selectively deamidated gliadin peptides. Clin Chem. 215. Malamut G, Cellier C.  Refractory celiac disease. Gastroenterol
2001;47(11):2023–8. Clin N Am. 2019;48(1):137–44.
199. Husby S, Murray JA, Katzka DA. AGA clinical practice update 216. Catassi C, Fabiani E, Iacono G, D’Agate C, Francavilla R, Biagi
on diagnosis and monitoring of celiac disease-changing utility of F, et  al. A prospective, double-blind, placebo-controlled trial to
serology and histologic measures: expert review. Gastroenterology. establish a safe gluten threshold for patients with celiac disease.
2019;156(4):885–9. Am J Clin Nutr. 2007;85(1):160–6.
200. Villalta D, Alessio MG, Tampoia M, Tonutti E, Brusca I, Bagnasco 217. Hischenhuber C, Crevel R, Jarry B, Maki M, Moneret-Vautrin
M, et al. Testing for IgG class antibodies in celiac disease patients DA, Romano A, et al. Review article: safe amounts of gluten for
with selective IgA deficiency. A comparison of the diagnostic patients with wheat allergy or coeliac disease. Aliment Pharmacol
accuracy of 9 IgG anti-tissue transglutaminase, 1 IgG anti-gliadin Ther. 2006;23(5):559–75.
and 1 IgG anti-deaminated gliadin peptide antibody assays. Clin 218. Elli L, Bascunan K, di Lernia L, Bardella MT, Doneda L, Soldati
Chim Acta. 2007;382(1–2):95–9. L, et al. Safety of occasional ingestion of gluten in patients with
201. Villalta D, Tonutti E, Prause C, Koletzko S, Uhlig HH, Vermeersch celiac disease: a real-life study. BMC Med. 2020;18(1):42.
P, et  al. IgG antibodies against deamidated gliadin peptides for 219. van der Fels-Klerx HJ, Smits N, Bremer M, Schultink JM, Nijkamp
diagnosis of celiac disease in patients with IgA deficiency. Clin MM, Castenmiller J, et al. Detection of gluten in d­ uplicate por-
Chem. 2010;56(3):464–8. tions to determine gluten intake of celiac disease patients on a
202. Gidrewicz D, Potter K, Trevenen CL, Lyon M, Butzner gluten free diet. Br J Nutr. 2020;125(9):1051–7.
JD.  Evaluation of the ESPGHAN celiac guidelines in a 220. Weisbrod VM, Silvester JA, Raber C, Suslovic W, Coburn SS,
North American pediatric population. Am J Gastroenterol. Raber B, et al. A quantitative assessment of gluten cross-contact in
2015;110(5):760–7. the school environment for children with celiac disease. J Pediatr
203. Badizadegan K, Vanlandingham DM, Hampton W, Thompson Gastroenterol Nutr. 2020;70(3):289–94.
KM.  Value of biopsy in a cohort of children with high-titer 221. Myleus A, Reilly NR, Green PHR. Rate, risk factors, and outcomes
celiac serologies: observation of dynamic policy differences of nonadherence in pediatric patients with celiac disease: a sys-
between Europe and North America. BMC Health Serv Res. tematic review. Clin Gastroenterol Hepatol. 2020;18(3):562–73.
2020;20(1):962. 222. Wolf RL, Lebwohl B, Lee AR, Zybert P, Reilly NR, Cadenhead
204. Gustafsson I, Repo M, Popp A, Kaukinen K, Hiltunen P, Arvola J, et al. Hypervigilance to a gluten-free diet and decreased quality
T, et  al. Prevalence and diagnostic outcomes of children with of life in teenagers and adults with celiac disease. Dig Dis Sci.
duodenal lesions and negative celiac serology. Dig Liver Dis. 2018;63(6):1438–48.
2020;52(3):289–95. 223. Ting A, Katz T, Sutherland R, Liu V, Tong CW, Gao Y, et  al.
205. Volta U, Caio G, Boschetti E, Giancola F, Rhoden KJ, Ruggeri E, Evaluating the dietary intakes of energy, macronutrients, sugar,
et  al. Seronegative celiac disease: shedding light on an obscure fiber, and micronutrients in children with celiac disease. J Pediatr
clinical entity. Dig Liver Dis. 2016;48(9):1018–22. Gastroenterol Nutr. 2020;71(2):246–51.
206. Leonard MM, Lebwohl B, Rubio-Tapia A, Biagi F. AGA clinical 224. Nestares T, Martin-Masot R, Labella A, Aparicio VA, Flor-­
practice update on the evaluation and management of seronegative Alemany M, Lopez-Frias M, et  al. Is a gluten-free diet enough
enteropathies. Gastroenterology. 2020;160(1):437–44. to maintain correct micronutrients status in young patients with
207. Aziz I, Evans KE, Hopper AD, Smillie DM, Sanders DS. A pro- celiac disease? Nutrients. 2020;12(3):844.
spective study into the aetiology of lymphocytic duodenosis. 225. Di Nardo G, Villa MP, Conti L, Ranucci G, Pacchiarotti C,
Aliment Pharmacol Ther. 2010;32(11–12):1392–7. Principessa L, et al. Nutritional deficiencies in children with celiac
548 S. Guandalini and V. Discepolo

disease resulting from a gluten-free diet: a systematic review. celiac disease on a gluten-free diet. J Pediatr Gastroenterol Nutr.
Nutrients. 2019;11(7):1588. 2017;65(3):267–9.
226. Raehsler SL, Choung RS, Marietta EV, Murray JA. Accumulation 239. Comino I, Fernandez-Banares F, Esteve M, Ortigosa L, Castillejo
of heavy metals in people on a gluten-free diet. Clin Gastroenterol G, Fambuena B, et  al. Fecal gluten peptides reveal limitations
Hepatol. 2018;16(2):244–51. of serological tests and food questionnaires for monitoring
227. Runde J, Mears M, Guandalini S, Jericho H. A narrow window: gluten-free diet in celiac disease patients. Am J Gastroenterol.
booming gluten-free market and fostering healthy dietary hab- 2016;111(10):1456–65.
its in children with celiac disease. J Pediatr Gastroenterol Nutr. 240. Silvester JA, Comino I, Kelly CP, Sousa C, Duerksen DR,
2020;71(4):533–5. Group DBS.  Most patients with celiac disease on gluten-free
228. Sansotta N, Guandalini S, Romano S, Amirikian K, Cipolli M, diets consume measurable amounts of gluten. Gastroenterology.
Tridello G, et al. The gluten free diet’s impact on growth in chil- 2020;158(5):1497–9. e1
dren with celiac disease in two different countries. Nutrients. 241. Stefanolo JP, Talamo M, Dodds S, de la Paz Temprano M, Costa
2020;12(6):1547. AF, Moreno ML, et  al. Real-world gluten exposure in patients
229. Wiech P, Chmiel Z, Bazalinski D, Salacinska I, Bartosiewicz with celiac disease on gluten-free diets, determined from glia-
A, Mazur A, et  al. The relationship between body composition din immunogenic peptides in urine and fecal samples. Clin
and a gluten free diet in children with celiac disease. Nutrients. Gastroenterol Hepatol. 2020;19(3):484–91.
2018;10(11):1817. 242. Alhassan E, Yadav A, Kelly CP, Mukherjee R. Novel nondietary
230. Ghiselli A, Bizzarri B, Gaiani F, Semeraro F, Iuliano S, Di therapies for celiac disease. Cell Mol Gastroenterol Hepatol.
Mario F, et al. Growth changes after gluteen free diet in pediatric 2019;8(3):335–45.
celiac patients: a literature-review. Acta Biomed. 2018;89(9-S): 243. Syage JA, Murray JA, PHR G, Khosla C. Latiglutenase improves
5–10. symptoms in seropositive celiac disease patients while on a
231. Pinto-Sanchez MI, Causada-Calo N, Bercik P, Ford AC, Murray gluten-­free diet. Dig Dis Sci. 2017;62(9):2428–32.
JA, Armstrong D, et  al. Safety of adding oats to a gluten-free 244. Konig J, Holster S, Bruins MJ, Brummer RJ. Randomized clini-
diet for patients with celiac disease: systematic review and meta-­ cal trial: effective gluten degradation by Aspergillus niger-derived
analysis of clinical and observational studies. Gastroenterology. enzyme in a complex meal setting. Sci Rep. 2017;7(1):13100.
2017;153(2):395–409. e3 245. Smecuol E, Constante M, Temprano MP, Costa AF, Moreno ML,
232. Sansotta N, Amirikian K, Guandalini S, Jericho H. Celiac disease Pinto-Sanchez MI, et al. Effect of Bifidobacterium infantis NLS
symptom resolution: effectiveness of the gluten-free diet. J Pediatr super strain in symptomatic coeliac disease patients on long-­
Gastroenterol Nutr. 2018;66(1):48–52. term gluten-free diet  – an exploratory study. Benefic Microbes.
233. Sansotta N, Guandalini S, Amirikian K, Jericho H. Predictors for 2020;11(6):527–34.
poor response in gastrointestinal and extra-intestinal symptoms in 246. McCarville JL, Nisemblat Y, Galipeau HJ, Jury J, Tabakman R,
a celiac population. Gastroenterology. 2017;152(5):S269. Cohen A, et  al. BL-7010 demonstrates specific binding to glia-
234. Sansotta N, Guandalini S, Amirikian K, Jericho H. Celiac disease din and reduces gluten-associated pathology in a chronic mouse
symptom resolution: effectiveness of the gluten free diet. J Pediatr model of gliadin sensitivity. PLoS One. 2014;9(11):e109972.
Gastroenterol Nutr. 2017;66(1):48–52. 247. Leffler DA, Kelly CP, Green PH, Fedorak RN, DiMarino A,
235. Ghazzawi Y, Rubio-Tapia A, Murray JA, Absah I. Mucosal heal- Perrow W, et  al. Larazotide acetate for persistent symptoms of
ing in children with treated celiac disease. J Pediatr Gastroenterol celiac disease despite a gluten-free diet: a randomized controlled
Nutr. 2014;59(2):229–31. trial. Gastroenterology. 2015;148(7):1311–9. e6
236. Vecsei E, Steinwendner S, Kogler H, Innerhofer A, Hammer K, 248. Kapoerchan VV, Wiesner M, Hillaert U, Drijfhout JW, Overhand
Haas OA, et  al. Follow-up of pediatric celiac disease: value of M, Alard P, et al. Design, synthesis and evaluation of high-affinity
antibodies in predicting mucosal healing, a prospective cohort binders for the celiac disease associated HLA-DQ2 molecule. Mol
study. BMC Gastroenterol. 2014;14:28. Immunol. 2010;47(5):1091–7.
237. Leonard MM, Weir DC, DeGroote M, Mitchell PD, Singh P, 249. ImmusanT Discontinues Phase 2 Clinical Trial for
Silvester JA, et al. Value of IgA tTG in predicting mucosal recov- Nexvax2® in Patients With Celiac Disease [Webpage].
ery in children with celiac disease on a gluten-free diet. J Pediatr 2019 [Available from: https://www.globenewswire.com/
Gastroenterol Nutr. 2017;64(2):286–91. n e w s -­r e l e a s e / 2 0 1 9 / 0 6 / 2 5 / 1 8 7 4 1 0 8 / 0 / e n / I m m u s a n T-­
238. Koletzko S, Auricchio R, Dolinsek J, Gillett P, Korponay-Szabo I, Discontinues-­Phase-­2-­Clinical-­Trial-­for-­Nexvax2-­in-­Patients-­
Kurppa K, et al. No need for routine endoscopy in children with With-­Celiac-­Disease.html

View publication stats

You might also like