Home Medicine Gut microbiome investigation in celiac disease: from methods to its pathogenetic role
Article Open Access

Gut microbiome investigation in celiac disease: from methods to its pathogenetic role

  • Lucia Sacchetti EMAIL logo and Carmela Nardelli
Published/Copyright: September 9, 2019

Abstract

Our body is inhabited by a variety of microbes (microbiota), mainly bacteria, that outnumber our own cells. Until recently, most of what we knew about the human microbiota was based on culture methods, whereas a large part of the microbiota is uncultivable, and consequently previous information was limited. The advent of culture-independent methods and, particularly, of next-generation sequencing (NGS) methodology, marked a turning point in studies of the microbiota in terms of its composition and of the genes encoded by these microbes (microbiome). The microbiome is influenced predominantly by environmental factors that cause a large inter-individual variability (~20%) being its heritability only 1.9%. The gut microbiome plays a relevant role in human physiology, and its alteration (“dysbiosis”) has been linked to a variety of inflammatory gut diseases, including celiac disease (CD). CD is a chronic, immune-mediated disorder that is triggered by both genetic (mainly HLA-DQ2/DQ8 haplotypes) and environmental factors (gluten), but, in recent years, a large body of experimental evidence suggested that the gut microbiome is an additional contributing factor to the pathogenesis of CD. In this review, we summarize the literature that has investigated the gut microbiome associated with CD, the methods and biological samples usually employed in CD microbiome investigations and the putative pathogenetic role of specific microbial alterations in CD. In conclusion, both gluten-microbe and host-microbe interactions drive the gluten-mediated immune response. However, it remains to be established whether the CD-associated dysbiosis is the consequence of the disease, a simple concomitant association or a concurring causative factor.

Introduction

The human body is inhabited by a variety of microbes, bacteria and, to a lesser extent, by archaea, fungi and viruses [1]. The number of bacteria and other microbes that we host outnumber our own cells. Indeed, the first estimated microbial to human cells was 10:1 [2], which is probably an overestimate depending on the proportion of bacteria in our guts, which stretches from the mouth to the anus, and from the more or less good estimate of the number of human cells. In fact, a recent study suggested a ratio of 1.3:1 microbial to human cells [3]. Notably, there is an average of 30 trillion (1012) human cells and 39 trillion bacteria in a man aged between 20 and 30 years, weighing 70 kg and 1.70 m tall [3]. The human microbiota is the ecological community of symbiotic, commensal and pathogenic microorganisms (the microbial taxa) that inhabit the surfaces and specific niches of our organism (gut, skin, mouth, etc.), whereas, the term human “microbiome” indicates the catalog of genes encoded by these microbes [4] (Table 1). In the past, most of what we knew about the human microbiota was based on culture methods, but it was estimated that as much as 20%–60% of human microbes are uncultivable [5], [6], [7], therefore previous information was limited. It was only after the development of culture-independent methods and, particularly, in the early 2000s, of next-generation sequencing (NGS) methodology [8], together with the development of powerful computational tools and subsequently of “omics” technologies (Table 1), that we were able to efficiently catalog microbes and their genes, and to establish the impact of the host-microbe interaction on human metabolism [9], [10], [11]. Microbiome changes have been linked to many complex immune-mediate diseases, among which celiac disease (CD) [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27].

Table 1:

Main glossary in microbiome studies.

Human microbiota: The trillions symbiotic microbial cells (bacteria, viruses or eukaryotes) inhabiting mucosal surfaces of our body
Human microbiome: The collective genes encoded by the human microbiota. The microbiome is different in the different niches of our body (i.e. duodenal microbiome is different from fecal or skin microbiomes)
Prevalent genes: Non-redundant genes present in the human gut microbiome
Common genes: Genes found in the gut microbiome of at least 50% of individuals
Gut microbiome: The catalog of 3.3 million non-redundant microbial genes present in the human gut
Gut common core: About 38% of the individual’s gut microbiome shared among different individuals
16S rRNA sequencing: Evaluation of the bacterial composition in a human niche (i.e. gut microbiome) based on the 16S rRNA marker (usually up to genus taxon level)
Metagenomics: Evaluation of the microbial composition in a human niche (i.e. gut microbiome) based on the whole genome sequencing of all microbes present (i.e. up to bacterial species and subspecies presence)
Metatranscriptomics: Evaluation of the microbial function in a human niche based on the whole RNA sequencing of all microbes present (i.e. gut metatranscriptomics)
“Omics” technologies: A set of methods used to investigate the interaction microbiome-host (i.e. metagenomics, metatranscriptomics, metaproteomics, metabolomics)
Dysbiosis: Imbalance of microbes inhabiting a human body niche

CD is a chronic, immune-mediated disorder that occurs at any age, and affects more women than men with a total prevalence of around 1% in Caucasians in Europe [28], [29]. CD involves both genetic and environmental factors [29]. The major contributing genes are those encoding the HLA-DQ2/DQ8 molecules [30] and, to a lesser extent, non-HLA genes [31]. Globally, genes account for 48% of the disease risk [31], [32], [33]. What is known so far about the pathogenesis of CD is that, in HLA-genetically predisposed individuals, wheat gluten proteins and proteins found in barley and rye, after ingestion, are modified by intestinal transglutaminase 2 (TG2) into highly immunogenic deamidated gluten peptides (DGP). The latter, after binding to HLA-DQ2/DQ8 molecules on dendritic cells (DCs), are presented to CD4+ T-cells thereby activating immune reactions (production of CD-specific antibodies, release of pro-inflammatory cytokines, and activation of intraepithelial lymphocytes) [34]. This succession of events induces inflammation and progressive histological changes in the small intestinal mucosa, namely an increase in intraepithelial lymphocytes (>25/100 enterocytes), crypt hyperplasia and villous atrophy [35]. Although gluten remains the major triggering factor of CD, several questions remain unanswered. Why does CD develop at any age even years after the introduction of gluten in the diet [36]? Why has the prevalence of CD increased over time in the Western world, although gluten-containing food consumption has not increased [37]? Why do all HLA DQ2/DQ8 genetically predisposed individuals not develop the disease [38]? Conflicting results have recently been reported regarding the influence on the CD risk of the timing and the amount of gluten consumption during infancy, likely due to different study designs [39], [40]. Furthermore, although a gluten-free diet (GFD) improves mucosal lesions in CD patients, it does not correct the intestinal immune response [41]. The aforementioned observations suggest that other pathogenic factors are involved in the onset of CD. We and others reported gut dysbiosis in adults and/or children affected by CD but not in controls, which indicates that the gut microbiome is an additional contributing factor to CD pathogenesis [12], [13], [14], [15], [16], [17], [19], [20], [21], [22], [23], [24], [25], [26], [27].

The gut microbiome, general features and its role in human physiology

Within the framework of the Human Microbiome Project, tissue samples from multiple body sites (gastrointestinal tract, mouth, vagina, skin, etc.) of 250 “normal” volunteers were analyzed to determine whether the monitoring or manipulation of the human microbiome could improve human health [42]. Subsequently, the study by Qin et al. [9], in which they examined fecal samples of 124 European individuals, was crucial in establishing the minimal gut metagenome (Table 1) and the minimal gut “bacterial” genome in terms of functions encoded by this gene set. In particular, the latter study [9], which was part of the Metagenomics of the Human Intestinal Tract (MetaHIT) Consortium Project established a catalog of 3.3 million non-redundant genes (prevalent genes) in the gut microbiome, most of which were present in only a few individuals (<20%), whereas 294,110 genes were considered “common genes” being found in at least 50% of individuals [9] (Table 1). In detail, each individual carries in their gut ~536,112 prevalent microbial genes among which ~204,056 are common genes, which indicates that about 38% of an individual’s gut microbiome is shared (gut common core) (Table 1) [9].

A comparison between the human genome (~22,000 genes) [43] and diversity (~99.9% identity between individuals) [44] with the microbial genes catalog (human gut microbiome ~3.3 million non-redundant genes) [9] and diversity (80%–90% diversity between individuals) [45] suggests that to study the microbiome together to human genome is mandatory to carry out “personalized medicine”.

As reported in Table 2, the gut microbiome plays relevant roles in human physiology: it produces vitamins, contributes to food digestion, preserves the structural integrity of the gut mucosal barrier thereby impeding the colonization of pathogens, participates in the gut-brain axis communication and primes the immune system thereby helping to maintain a healthy status [46]. However, it is not easy to define the “gut health microbiome”. In fact, the microbiome composition is influenced by several factors: host genetics, diet, drugs, lifestyle and other environmental factors. Rothschild et al. [47] recently examined approximately 1000 healthy individuals of different ancestral origin and found no significant differences between ancestries in terms of microbiome composition, which suggested that the microbiome is not closely associated with host genetics. The same authors estimated that the microbiome heritability weight was only 1.9% in a dataset of 2252 twins [48]. By contrast, significant microbiome similarities were found in first-degree relative pairs and in pairs of genetically unrelated individuals who reported sharing a household [47]. In detail, about 20% of inter-person microbiome variability was associated with multiple environmental factors (diet, drugs, physical activity, etc.) and the body mass index [47]. Globally, microbiome composition is predominantly shaped by non-genetic factors that cause a large intra- and inter-individual variability, which makes it difficult to define what is a “healthy gut microbiota”. However, although each individual is characterized by a specific combination of bacterial taxa, gut microbiota functions are highly preserved. Therefore, from a clinical point of view, any microbial pattern associated with a healthy host status could be considered “healthy” [49].

Table 2:

Role of gut microbiota in human physiology.

Gut microbiota function Metabolites produced by gut microbiota and their impact on the host
Metabolism Fermented end products: SCFAsa; microbes (Roseburia hominis and Fecalibacterium prausnitzi [Firmicutes], etc.); energy substrate for the host; ATP production in colonocytes
Host energy harvesting (i.e. unabsorbed starch and soluble dietary fiber fermentation)
Production of vitamins and acetylcholine Vitamins K, B (B5, B12) and choline metabolites; microbes (Bacteroides fragilis, Eubacterium lentum, Serratia marcescens, etc.); lipid metabolism; energy production; enzymatic cofactor; nervous system functioning
Bile acids metabolism
Production of secondary bile acids; microbes (Clostridium perfrigens and C. scindens); glucose and energy metabolism; antimicrobial effects
Gut barrier integrity and permeability SCFAsa; microbes (R. hominis and F. prausnitzi [Firmicutes], etc.); downregulation of pathogens virulence factors and modulation of pH; nutrient competition with pathogens
Colonization resistance
Host immune maturation and function Polyamines (putrescine, spermidine, spermine); indole derivatives; Microbes (B. fragilis [Bacteroidetes], Bifidobacterium infantis [Actinobacteria] and Firmicutes, etc.); enhancement of the intestinal barrier integrity (by stimulation of intercellular junction proteins synthesis), inhibition of macrophage activation and tight regulation of Treg/Th17 ratio
Aberrant immune-inflammatory response prevention
Modulation and function of the enteric nervous system (ENS) SCFAsa; Microbes (R. hominis and F. prausnitzi [Firmicutes], etc.); recognition and reaction of ENSa (via TLR4a) to the G(−) bacterial LPSa; gut-brain axis: bidirectional communication of ENS with CNSa regulates a variety of gastrointestinal functions via vagal pathways (exocrine/endocrine secretions, gut motility, immune-inflammatory reactions, etc.)
Intestinal homeostasis
  1. aSCFAs, short chain fatty acids: butyrate, propionate, acetate and pentanoate; ENS, enteric nervous system; CNS, central nervous system; TLR4, toll like receptor 4; LPS, lipopolysaccharide.

In support of the relevance of microbiome functions, a recent study identified metagenomically abundant organisms that were inactive or dormant in the gut with being little or not expressed [50], which indicates that disease-specific microbial alterations can sometimes be detectable only at the transcriptional level. Globally, it is now evident that, besides the microbial composition (metagenomics) of the gut, also its activity (metatranscriptomics) should be evaluated (Table 1) in order to explore the role of the microbiome in human health and disease [51].

Gut microbiome profiles in controls and in patients affected by CD

As mentioned, the gut microbiota is composed of several types of microrganisms, namely bacteria, archea, fungi and viruses, among which bacteria are the most abundant [1]. To date, most of the microbiota studies conducted in patients affected by CD have been bacteria-centric [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], whereas the mycobiome and virome have also been studied as implicated in the pathogenesis of CD [52], [53], [54]. Currently, the 16S rRNA gene marker evaluation and NGS (see Supplementary material) are the most widely used techniques with which to explore the abundance and diversity of bacterial taxa composition (from phylum to class, order, family, genus) present in a human body niche or sample.

A few phyla dominate the healthy human gut: Firmicutes, Bacteroidetes, Actinobacteria, Fusobacteria, Proteobacteria and Verrucomicrobia. The first two phyla constitute about 90% of the gut microbiota [55]. Clostridium, Lactobacillus and Enterococcus are the most abundant Firmicutes, Bacteroides and Prevotella are the most abundant Bacteroidetes, whereas Bifidobacterium is the most abundant Actinobacteria [55]. Microbiota composition differs in the various parts of the gastrointestinal tract, which is affected by pH, host secretion, substrate availability, etc. thereby resulting in a craniocaudal progressive decrease of aerobic and an increase in strictly anaerobic bacterial species [56]. Consequently, the gut microbiome profile associated with a healthy condition and/or with an inflammatory gut disease, including CD, depends on the specific niche or sample investigated, as well as on the all other factors influencing microbiota variability (Table 3).

Table 3:

Gut dysbiotic features observed in active CD patients.

Biological sample CD associated dysbiosis References
Feces ↑ Gram (−)/Gram (+) bacteria ratio

↓ Firmicutes (Lactobacillus spp., Faecalibacterium prausnitzii, Clostridium spp.)

↓ Actinobacteria (Bifidobacterium spp.)

↑ Bacteroidetes (Bacteroides spp.)

↑ Proteobacteria (E. coli)

↑ Firmicutes (Staphylococcus spp.)
Sanz et al. [12]

Di Cagno et al. [13]

Collado et al. [14]

De Palma et al. [15]

Quagliariello et al. [16]

Olivares et al. [17]
Duodenal mucosa ↑ Gram (−) bacteria

↓ Firmicutes (Lactobacillus spp., Streptococcus spp.)

↓ Bacteroidetes (Prevotella spp.)

↑ Proteobacteria (Neisseria spp., E. coli)
Nadal et al. [19]

Collado et al. [14]

Schippa et al. [20]

Di Cagno et al. [21]

Nistal et al. [22]

Wacklin et al. [23]

Sánchez et al. [24]

D’Argenio et al. [25]

Iaffaldano et al. [26]
Saliva ↓ Bacteroidetes

↓ Fusobacteria

↑ Actinobacteria
Tian et al. [27]
Oropharingeal swab ↓ Bacteroidetes

↓ Fusobacteria

↑ Actinobacteria (Actimomyces spp.)

↑ Proteobacteria (Nf)
Iaffaldano et al. [26]

Oral microbiome

Oral microbiota is the second most complex and abundant microbial community in the human body after the colon [42]. The microbiome associated with CD patients has also been investigated in saliva and oropharyngeal swabs, which are easy to obtain and to study [26], [27]. The rationale of studying the oral microbiome is that gluten is first processed in the mouth, consequently the oral microbiome might have an impact on the immunogenic peptides produced after this first part of the digestion process. Recently, Tian et al. [27] reported an increased abundance of Actinobacteria and a reduced presence of Bacteroidetes and Fusobacteria phyla in the oral microbiota of refractory CD patients. They hypothesized that the higher levels of Lactobacilli they found in CD patients than in healthy individuals caused the higher CD-associated gluten-degrading activity observed in CD patients [27]. As shown in Figure 1, we recently investigated the oropharyngeal and duodenal microbiomes of CD patients to evaluate if these two niches have some CD-associated microbial alterations in common [26]. Intriguingly, we found that the duodenum and oropharynx shared similar microbiome profiles, characterized by a high abundance of the Proteobacteria phylum, and Neisseria species, mostly accounting for this abundance. Our results suggest a continuum of microbiome in active CD patients from mouth to duodenum (Figure 1).

Figure 1: Microbiome taxonomic composition in controls (C), in active (a-CD) and in GFD celiac patients.
The bar plots show the relative abundance (%) at phylum (A, B) and genus (C, D) levels in duodenal and oropharyngeal samples (A, C and B, D, respectively). Phyla and genera with an abundance greater than 1% in at least one study group are reported. Proteobacteria was the most abundant phylum in both duodenal and oropharyngeal samples of a-CD patients (A, B). The Neisseria genus was significantly more abundant in duodenum (p<0.005) and in oropharynx (p<0.001) of a-CD patients than in controls and GFD patients (C and D, respectively). The microbial profiles obtained in duodenal and oropharyngeal samples in each a-CD patient (E) had a very similar composition, indeed, the Proteobacteria phylum was most abundant phylum in duodenum and oropharynx niches (45% and 52%, respectively). The latter finding suggests a continuum from mouth to duodenum in the microbiome of a-CD patients (E). Error bars indicate standard error. Asterisks refer to taxa that differed significantly among three groups. *p<0.05; **p<0.005; ***p<0.001. (Modified from Refs. [25] and [26] with permission.)
Figure 1:

Microbiome taxonomic composition in controls (C), in active (a-CD) and in GFD celiac patients.

The bar plots show the relative abundance (%) at phylum (A, B) and genus (C, D) levels in duodenal and oropharyngeal samples (A, C and B, D, respectively). Phyla and genera with an abundance greater than 1% in at least one study group are reported. Proteobacteria was the most abundant phylum in both duodenal and oropharyngeal samples of a-CD patients (A, B). The Neisseria genus was significantly more abundant in duodenum (p<0.005) and in oropharynx (p<0.001) of a-CD patients than in controls and GFD patients (C and D, respectively). The microbial profiles obtained in duodenal and oropharyngeal samples in each a-CD patient (E) had a very similar composition, indeed, the Proteobacteria phylum was most abundant phylum in duodenum and oropharynx niches (45% and 52%, respectively). The latter finding suggests a continuum from mouth to duodenum in the microbiome of a-CD patients (E). Error bars indicate standard error. Asterisks refer to taxa that differed significantly among three groups. *p<0.05; **p<0.005; ***p<0.001. (Modified from Refs. [25] and [26] with permission.)

Duodenal microbiome

Interestingly, despite the different chemical and physical conditions present in the upper and lower tracts of the intestine, and different methodological approaches, some CD-associated fecal dysbiosis profiles were also observed in the duodenum of both children and adult CD patients compared to controls [13], [19], [20], [21], [22], [23], [24]. In fact, there was an increased abundance of species belonging to the Proteobacteria phylum (Escherichia coli and Neisseria) and a decreased abundance of Firmicutes (Lactobacillus, Streptococcus) and Actinobacteria (Bifidobacteria) phyla [14], [19], [20], [21], [22], [23], [24].

As shown in Figure 1, we recently characterized the duodenal microbiome profiles in active and GFD adult CD patients and controls using 16S rRNA sequencing, and, in agreement with above data, which were mostly obtained using diverse methodological approaches, found that Proteobacteria was the most abundant phylum, and Firmicutes and Actinobacteria were the least abundant phyla in the microbiome profiles of active CD patients [25], [26]. Furthermore, we highlighted that members of the Neisseria genus (β-Proteobacteria class), later identified as Neisseria flavescens (CD-Nf) by culture-based methods followed by mass spectrometry, were significantly more abundant in active CD patients than in GFD patients and control individuals [25], [26].

In conclusion, although some authors did not find any statistically significant difference between the duodenal microbiomes of CD patients and controls, probably due to the low resolution method used and/or to the low number of subjects studied [57], [58], [59], most of the scientific evidence is in line with a duodenal dysbiosis profile in patients affected by CD.

Fecal microbiome

Feces have been widely used to investigate gut CD dysbiosis in children. The early gut microbiota studies were based on culture methods combined or not with other technologies (denaturing gradient gel electrophoresis [DGGE], fluorescence in situ hybridization [FISH], temporal temperature gradient gel electrophoresis [TTGE] and real time-polymerase chain reaction [RT-PCR]) and consequently did not identify all microbial taxa. Thanks to the advent of NGS, it is now possible to characterize all the bacteria in gut samples (Supplementary Figure 1). Most studies revealed an increase in Gram (−) vs. Gram(+) bacteria ratio, an increase in E. coli and Bacteroides, and a decrease in Lactobacillus and Bifidobacteria in feces from CD patients compared to controls [12], [13], [14], [15], [16], [17] (Table 3). Furthermore, Staphylococcus epidermidis was more abundant in stools from CD children than in controls [14], [18], [24] (Table 3).

The putative pathogenic role of CD-associated dysbiosis

Numerous studies focused on the putative causal role of specific bacterial groups in the pathogenesis of CD [18], [25], [27], [54], [60], [61], [62], [63], [64], [65], [66]. Below, we summarize results that suggest the putative pathogenic role of specific microbes in CD.

Host immune-microbiota interaction

A higher abundance of virulent genes and in vitro production of pro-inflammatory cytokines was found in bacterial strains isolated from CD patients than in controls, as in the case of Staphylococcus spp. [18], E. coli [60], Bacteroides fragilis [61] and N. flavescens (Nf) [25]. In particular, using whole genome sequencing, we found that the gene system of iron acquisition differed between CD- and control-associated Nf. In fact, CD-Nf had the hmbR gene, which is usually present in the pathogenic Neisseria species Neisseria meningitides, and lacked the hpuA/B and tbpA/B genes, present in commensal Nf strains [25]. Furthermore, CD-Nf induced inflammatory phenotypes in ex-vivo mucosal explants, and in both human and murine DCs [25] (Figure 2). We also observed statistically significant lower mitochondrial activity, accompanied by lower ATP production and by higher oxidative stress in CD-Nf-infected Caco-2 cells than in untreated Caco-2 cells [62]. Based on the aforementioned metabolic alterations, we hypothesized that the CD-Nf that we isolated from the duodenum of CD patients, could contribute to the pathogenesis of CD or to disease exacerbation.

Figure 2: In vitro inflammation effects of CD-associated Nf.
(A) Nf isolated from celiac disease (CD) patients induced a pro-inflammatory phenotype on dendritic cells. Filtered bacterial lysates from Nf, isolated from five active celiac patients (CD-Nf 1, 2, 3, 7, and 8), promoted the production of pro-inflammatory cytokines by dendritic cells (DCs). CD11c+DCs isolated from the spleens of C57B6 mice were cultured for 48 h in the presence of retinoic acid (RA) and transforming growth factor-β (TGF-β) with filtered Nf lysates from three active CD patients. All three tested bacterial lysates promoted the production of interleukin (IL)-12p40 by murine DCs, whereas untreated (* on horizontal bars), lipopolysaccharide (LPS)-treated (**), and LGG-treated DCs did not. (B) Human monocyte-derived DCs were cultured for 48 h in the presence of RA and TGF-β with filtered Nf lysates isolated from active CD patients. Tumor necrosis factor-α (TNF-α) levels were higher in supernatants of human DCs treated with Nf 1, Nf 7 and Nf 8 bacterial strains isolated from small intestinal biopsies of CD patients, and in those challenged with Nf 10 isolated from a pharyngeal swab of a control subject, than in those treated with bacterial LPS, used as a pro-inflammatory positive control (stars upon horizontal bars) and in untreated (UN) cells (p<0.0001 for each of the treatments). IL-12p40 levels were higher in supernatants of human DCs challenged with Nf 1, 2, 3 filtered lysates than in those treated with bacterial LPS (**) used as a proinflammatory positive control and in untreated (UN) cells (stars upon horizontal bars). Dot plots represent values from three independent experiments. Each experiment was performed in duplicate. A two-way analysis of variance was performed to check for inter-group differences. *p<0.05, **p<0.01, ***p<0.001, and ****p<0.0001. (C) Immunofluorescence analysis showed that in ex-vivo mucosal explants from control subjects the CD-Nf challenge enhanced the epithelial expression of the inflammation marker HLA-DR in villus enterocytes, in the basal cytoplasmatic compartment as well as on the brush border and basolateral membranes. The HLA-DR scoring was graded from absent to very strong: 0–3 and measured the increase of HLA-DR expression. (Modified from Ref. [25] with permission.)
Figure 2:

In vitro inflammation effects of CD-associated Nf.

(A) Nf isolated from celiac disease (CD) patients induced a pro-inflammatory phenotype on dendritic cells. Filtered bacterial lysates from Nf, isolated from five active celiac patients (CD-Nf 1, 2, 3, 7, and 8), promoted the production of pro-inflammatory cytokines by dendritic cells (DCs). CD11c+DCs isolated from the spleens of C57B6 mice were cultured for 48 h in the presence of retinoic acid (RA) and transforming growth factor-β (TGF-β) with filtered Nf lysates from three active CD patients. All three tested bacterial lysates promoted the production of interleukin (IL)-12p40 by murine DCs, whereas untreated (* on horizontal bars), lipopolysaccharide (LPS)-treated (**), and LGG-treated DCs did not. (B) Human monocyte-derived DCs were cultured for 48 h in the presence of RA and TGF-β with filtered Nf lysates isolated from active CD patients. Tumor necrosis factor-α (TNF-α) levels were higher in supernatants of human DCs treated with Nf 1, Nf 7 and Nf 8 bacterial strains isolated from small intestinal biopsies of CD patients, and in those challenged with Nf 10 isolated from a pharyngeal swab of a control subject, than in those treated with bacterial LPS, used as a pro-inflammatory positive control (stars upon horizontal bars) and in untreated (UN) cells (p<0.0001 for each of the treatments). IL-12p40 levels were higher in supernatants of human DCs challenged with Nf 1, 2, 3 filtered lysates than in those treated with bacterial LPS (**) used as a proinflammatory positive control and in untreated (UN) cells (stars upon horizontal bars). Dot plots represent values from three independent experiments. Each experiment was performed in duplicate. A two-way analysis of variance was performed to check for inter-group differences. *p<0.05, **p<0.01, ***p<0.001, and ****p<0.0001. (C) Immunofluorescence analysis showed that in ex-vivo mucosal explants from control subjects the CD-Nf challenge enhanced the epithelial expression of the inflammation marker HLA-DR in villus enterocytes, in the basal cytoplasmatic compartment as well as on the brush border and basolateral membranes. The HLA-DR scoring was graded from absent to very strong: 0–3 and measured the increase of HLA-DR expression. (Modified from Ref. [25] with permission.)

In addition to bacteria, also viruses are involved in the pathogenesis of CD [54]. In fact, although reovirus infection does not cause symptoms it could interact with the immune system and affect its response to food antigens thereby promoting a pro-inflammatory rather than a regulatory response to them [54]. The latter finding was supported by the break of tolerance to gluten in mice infected with reovirus and by antibody titers against reovirus in patients with CD vs. controls [54].

Gluten-microbiota interaction

As mentioned, the oral cavity is the first part of the gastrointestinal tract where wheat-derived gliadins may be substrates for oral microbial proteases originating from the bacteria naturally residing in the mouth [63]. In particular, Rothia mucilaginosa and Rothia aeria [64], [65] and Lactobacilli spp. [27], which are highly active towards gluten have been identified in the oral microbiota [27]. Other gluten-degrading organisms, including the immunogenic domains of gluten, are Actinomyces odontolyticus, Streptococcus mitis, Neisseria mucosa and Capnocytophaga sputigena [65]. Increased proteolytic activity against gluten has also been observed in the duodenum of CD patients in correlation with increased levels of Proteobacteria, including Pseudomonas [66]. Interestingly, in mice expressing CD risk genes, Pseudomonas aeruginosa elastase synergizes with gluten to induce more severe inflammation than that associated with mild villus blunting [66]. Moreover, microbial transglutaminase (mTG), an enzyme frequently used in food production, whose enzymatic properties are similar to those of human tissue transglutaminase (tTG), might be involved in the increase of CD [67]. In fact, mTG and gliadin are transported to the endoplasmic reticulum of enterocytes and mTG strongly localizes to the basolateral membrane and lamina propria, which indicates an antigenic interaction of mTG with cells of the immune system. Because mTG may also be released by bacteria (mainly those belonging to the Firmicutes phylum) within the intestinal microbiota, and also by external sources such as probiotics, mTG might play a role in the pathogenesis of CD [67], [68].

Concluding remarks

In conclusion, in vivo and in vitro studies definitively support an association between the gut microbiome and CD. Thus, both gluten-microbes and host-microbes interactions drive the gluten-mediated immune response. But, it remains to be established whether the CD-associated dysbiosis is the consequence of the disease, a simple concomitant association or a concurring causative factor. CD is a complex multifactorial disorder and there is no model system that summarizes its complexity. All current in vitro models, namely, human duodenal mucosal explants, immortalized cell lines (e.g. Caco2 cells), humanized mouse models, human intestinal organoids, have advantages and disadvantages (reviewed in ref. [69]). Recently, an intestine-on-a-chip system has been developed that forms polarized 3D-villus-like structures, and has basic functional properties and an intestinal microenvironment [70]. This is the device that most closely resembles the in vivo human intestine and, hopefully, might help to clarify the role of the microbiome in the pathogenesis of CD. Indeed, by combining CD patient-derived human-induced pluripotent stem cells and human intestinal organoids, it will be possible to create intestine-on-a-chip systems that include individual’s genetic background in order to implement also “personalized” therapeutic interventions.


Corresponding author: Prof. Lucia Sacchetti, CEINGE-Biotecnologie Avanzate SCarl, Via G. Salvatore 486, 80145 Naples, Italy; and Task Force on Microbiome Studies, Università degli Studi di Napoli Federico II and CEINGE-Biotecnologie Avanzate SCarl, Naples, Italy, Phone: 0039 081 3737827

Acknowledgments

The authors thank Jean Ann Gilder (Scientific Communication srl., Naples, Italy) for editing the text, and Vittorio Lucignano, CEINGE-Biotecnologie Avanzate, for technical assistance related to graphics.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Marchesi JR. Prokaryotic and eukaryotic diversity of the human gut. Adv Appl Microbiol 2010;72:43–62.10.1016/S0065-2164(10)72002-5Search in Google Scholar

2. Luckey TD. Introduction to intestinal microecology. Am J Clin Nutr 1972;25:1292–4.10.1093/ajcn/25.12.1292Search in Google Scholar PubMed

3. Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 2016;164:337–40.10.1016/j.cell.2016.01.013Search in Google Scholar PubMed

4. Lederberg J, McCray AT. ‘Ome sweet ‘Omics – a genealogical treasury of words. Scientist 2001;15:8.Search in Google Scholar

5. Pei Z, Bini EJ, Yang L, Zhou M, Francois F, Blaser MJ. Bacterial biota in the human distal esophagus. Proc Natl Acad Sci U S A 2004;101:4250–5.10.1073/pnas.0306398101Search in Google Scholar PubMed PubMed Central

6. Zhou X, Bent SJ, Schneider MG, Davis CC, Islam MR, Forney LJ. Characterization of vaginal microbial communities in adult healthy women using cultivation-independent methods. Microbiology 2004;150(Pt 8):2565–73.10.1099/mic.0.26905-0Search in Google Scholar PubMed

7. Aas JA, Paster BJ, Stokes LN, Olsen I, Dewhirst FE. Defining the normal bacterial flora of the oral cavity. J Clin Microbiol 2005;43:5721–32.10.1128/JCM.43.11.5721-5732.2005Search in Google Scholar PubMed PubMed Central

8. Metzker ML. Sequencing technologies – the next generation. Nat Rev Genet 2010;11:31–46.10.1038/nrg2626Search in Google Scholar PubMed

9. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010;464:59–65.10.1038/nature08821Search in Google Scholar PubMed PubMed Central

10. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods 2010;7:335–6.10.1038/nmeth.f.303Search in Google Scholar PubMed PubMed Central

11. Lepage P, Leclerc MC, Joossens M, Mondot S, Blottière HM, Raes J, et al. A metagenomic insight into our gut’s microbiome. Gut 2013;62:146–58.10.1136/gutjnl-2011-301805Search in Google Scholar PubMed

12. Sanz Y, Sánchez E, Marzotto M, Calabuig M, Torriani S, Dellaglio F. Differences in faecal bacterial communities in coeliac and healthy children as detected by PCR and denaturing gradient gel electrophoresis. FEMS Immunol Med Microbiol 2007;51:562–8.10.1111/j.1574-695X.2007.00337.xSearch in Google Scholar PubMed

13. Di Cagno R, Rizzello CG, Gagliardi F, Ricciuti P, Ndagijimana M, Francavilla R, et al. Different fecal microbiotas and volatile organic compounds in treated and untreated children with celiac disease. Appl Environ Microbiol 2009;75:3963–71.10.1128/AEM.02793-08Search in Google Scholar PubMed PubMed Central

14. Collado MC, Donat E, Ribes-Koninckx C, Calabuig M, Sanz Y. Specific duodenal and faecal bacterial groups associated with paediatric coeliac disease. J Clin Pathol 2009;62:264–9.10.1136/jcp.2008.061366Search in Google Scholar PubMed

15. De Palma G, Nadal I, Medina M, Donat E, Ribes-Koninckx C, Calabuig M, et al. Intestinal dysbiosis and reduced immunoglobulin-coated bacteria associated with coeliac disease in children. BMC Microbiol 2010;10:63.10.1186/1471-2180-10-63Search in Google Scholar PubMed PubMed Central

16. Quagliariello A, Aloisio I, Bozzi Cionci N, Luiselli D, D’Auria G, Martinez-Priego L, et al. Effect of bifidobacterium breve on the intestinal microbiota of coeliac children on a gluten free diet: a pilot study. Nutrients 2016;8. pii: E660.10.3390/nu8100660Search in Google Scholar PubMed PubMed Central

17. Olivares M, Walker AW, Capilla A, Benítez-Páez A, Palau F, Parkhill J, et al. Gut microbiota trajectory in early life may predict development of celiac disease. Microbiome 2018;6:36.10.1186/s40168-018-0415-6Search in Google Scholar PubMed PubMed Central

18. Sánchez E, Ribes-Koninckx C, Calabuig M, Sanz Y. Intestinal Staphylococcus spp. and virulent features associated with coeliac disease. J Clin Pathol 2012;65:830–4.10.1136/jclinpath-2012-200759Search in Google Scholar PubMed

19. Nadal I, Donat E, Ribes-Koninckx C, Calabuig M, Sanz Y. Imbalance in the composition of the duodenal microbiota of children with coeliac disease. J Med Microbiol 2007;56(Pt 12):1669–74.10.1099/jmm.0.47410-0Search in Google Scholar PubMed

20. Schippa S, Iebba V, Barbato M, Di Nardo G, Totino V, Checchi MP, et al. A distinctive ‘microbial signature’ in celiac pediatric patients. BMC Microbiol 2010;10:175.10.1186/1471-2180-10-175Search in Google Scholar PubMed PubMed Central

21. Di Cagno R, De Angelis M, De Pasquale I, Ndagijimana M, Vernocchi P, Ricciuti P, et al. Duodenal and faecal microbiota of celiac children: molecular, phenotype and metabolome characterization. BMC Microbiol 2011;11:219.10.1186/1471-2180-11-219Search in Google Scholar PubMed PubMed Central

22. Nistal E, Caminero A, Herrán AR, Arias L, Vivas S, de Morales JM, et al. Differences of small intestinal bacteria populations in adults and children with/without celiac disease: effect of age, gluten diet, and disease. Inflamm Bowel Dis 2012;18:649–56.10.1002/ibd.21830Search in Google Scholar PubMed

23. Wacklin P, Kaukinen K, Tuovinen E, Collin P, Lindfors K, Partanen J, et al. The duodenal microbiota composition of adult celiac disease patients is associated with the clinical manifestation of the disease. Inflamm Bowel Dis 2013;19:934–41.10.1097/MIB.0b013e31828029a9Search in Google Scholar PubMed

24. Sánchez E, Donat E, Ribes-Koninckx C, Fernández-Murga ML, Sanz Y. Duodenal-mucosal bacteria associated with celiac disease in children. Appl Environ Microbiol 2013;79:5472–9.10.1128/AEM.00869-13Search in Google Scholar PubMed PubMed Central

25. D’Argenio V, Casaburi G, Precone V, Pagliuca C, Colicchio R, Sarnataro D, et al. Metagenomics reveals dysbiosis and a potentially pathogenic N. flavescens strain in duodenum of adult celiac patients. Am J Gastroenterol 2016;111:879–90.10.1038/ajg.2016.95Search in Google Scholar PubMed PubMed Central

26. Iaffaldano L, Granata I, Pagliuca C, Esposito MV, Casaburi G, Salerno G, et al. Oropharyngeal microbiome evaluation highlights Neisseria abundance in active celiac patients. Sci Rep 2018;8:11047.10.1038/s41598-018-29443-1Search in Google Scholar PubMed PubMed Central

27. Tian N, Faller L, Leffler DA, Kelly CP, Hansen J, Bosch JA, et al. Salivary gluten degradation and oral microbial profiles in healthy individuals and celiac disease patients. Appl Environ Microbiol 2017;83. pii: e03330–16.10.1128/AEM.03330-16Search in Google Scholar PubMed PubMed Central

28. Mustalahti K, Catassi C, Reunanen A, Fabiani E, Heier M, McMillan S, et al. The prevalence of celiac disease in Europe: results of a centralized, international mass screening project. Ann Med 2010;42:587–95.10.3109/07853890.2010.505931Search in Google Scholar PubMed

29. Ludvigsson JF, Leffler DA, Bai JC, Biagi F, Fasano A, Green PH, et al. The Oslo definitions for coeliac disease and related terms. Gut 2013;62:43–52.10.1136/gutjnl-2011-301346Search in Google Scholar PubMed PubMed Central

30. Bourgey M, Calcagno G, Tinto N, Gennarelli D, Margaritte-Jeannin P, Greco L, et al. HLA related genetic risk for coeliac disease. Gut 2007;56:1054–9.10.1136/gut.2006.108530Search in Google Scholar PubMed PubMed Central

31. Izzo V, Pinelli M, Tinto N, Esposito MV, Cola A, Sperandeo MP, et al. Improving the estimation of celiac disease sibling risk by non-HLA genes. PLoS One 2011;6:e26920.10.1371/journal.pone.0026920Search in Google Scholar PubMed PubMed Central

32. Kuja-Halkola R, Lebwohl B, Halfvarson J, Wijmenga C, Magnusson PK, Ludvigsson JF. Heritability of non-HLA genetics in coeliac disease: a population-based study in 107 000 twins. Gut 2016;65:1793–8.10.1136/gutjnl-2016-311713Search in Google Scholar

33. Gutierrez-Achury J, Zhernakova A, Pulit SL, Trynka G, Hunt KA, Romanos J, et al. Fine mapping in the MHC region accounts for 18% additional genetic risk for celiac disease. Nat Genet 2015;47:577–8.10.1038/ng.3268Search in Google Scholar

34. Di Sabatino A, Corazza GR. Coeliac disease. Lancet 2009;373:1480–93.10.1016/S0140-6736(09)60254-3Search in Google Scholar

35. Marsh MN. Defining ‘coeliac’: Oslo Accord – or not? Gut 2013;62:1669–70.10.1136/gutjnl-2013-304632Search in Google Scholar PubMed

36. Vivas S, Ruiz de Morales JM, Fernandez M, Hernando M, Herrero B, Casqueiro J, et al. Age-related clinical, serological, and histopathological features of celiac disease. Am J Gastroenterol 2008;103:2360–5; quiz 2366.10.1111/j.1572-0241.2008.01977.xSearch in Google Scholar PubMed

37. Lohi S, Mustalahti K, Kaukinen K, Laurila K, Collin P, Rissanen H, et al. Increasing prevalence of coeliac disease over time. Aliment Pharmacol Ther 2007;26:1217–25.10.1111/j.1365-2036.2007.03502.xSearch in Google Scholar PubMed

38. Green PH, Cellier C. Celiac disease. N Engl J Med 2007;357: 1731–43.10.1056/NEJMra071600Search in Google Scholar PubMed

39. Crespo-Escobar P, Mearin ML, Hervás D, Auricchio R, Castillejo G, Gyimesi J, et al. The role of gluten consumption at an early age in celiac disease development: a further analysis of the prospective PreventCD cohort study. Am J Clin Nutr 2017;105: 890–6.10.3945/ajcn.116.144352Search in Google Scholar PubMed

40. Mårild K, Dong F, Lund-Blix NA, Seifert J, Barón AE, Waugh KC, et al. Gluten intake and risk of celiac disease: long-term follow-up of an at-risk birth cohort. Am J Gastroenterol 2019;114: 1307–14.10.14309/ajg.0000000000000255Search in Google Scholar PubMed PubMed Central

41. Lahdenperä A, Ludvigsson J, Fälth-Magnusson K, Högberg L, Vaarala O. The effect of gluten-free diet on Th1-Th2-Th3-associated intestinal immune responses in celiac disease. Scand J Gastroenterol 2011;46:538–49.10.3109/00365521.2011.551888Search in Google Scholar PubMed

42. NIH HMP Working Group, Peterson J, Garges S, Giovanni M, McInnes P, Wang L, et al. The NIH Human Microbiome Project. Genome Res 2009;19:2317–23.10.1101/gr.096651.109Search in Google Scholar PubMed PubMed Central

43. International Human Genome Sequencing Consortium. Finishing the euchromatic sequence of the human genome. Nature 2004;431:931–45.10.1038/nature03001Search in Google Scholar PubMed

44. Wheeler DA, Srinivasan M, Egholm M, Shen Y, Chen L, McGuire A, et al. The complete genome of an individual by massively parallel DNA sequencing. Nature 2008;452:872–6.10.1038/nature06884Search in Google Scholar PubMed

45. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The Human Microbiome Project. Nature 2007;449:804–10.10.1038/nature06244Search in Google Scholar PubMed PubMed Central

46. Kho ZY, Laf SK. The human gut microbiome – a potential controller of wellness and disease. Front Microbiol 2018;9:1835.10.3389/fmicb.2018.01835Search in Google Scholar PubMed PubMed Central

47. Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem T, Zeevi D, et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018;555:210–5.10.1038/nature25973Search in Google Scholar PubMed

48. Goodrich JK, Davenport ER, Clark AG, Ley RE. The relationship between the human genome and microbiome comes into view. Annu Rev Genet 2017;51:413–33.10.1146/annurev-genet-110711-155532Search in Google Scholar PubMed PubMed Central

49. Schmidt TS, Raes J, Bork P. The human gut microbiome: from association to modulation. Cell 2018;172:1198–215.10.1016/j.cell.2018.02.044Search in Google Scholar PubMed

50. Schirmer M, Franzosa EA, Lloyd-Price J, McIver LJ, Schwager R, Poon TW, et al. Dynamics of metatranscription in the inflammatory bowel disease gut microbiome. Nat Microbiol 2018;3:337–46.10.1038/s41564-017-0089-zSearch in Google Scholar PubMed PubMed Central

51. Abu-Ali GS, Mehta RS, Lloyd-Price J, Mallick H, Branck T, Ivey KL, et al. Metatranscriptome of human faecal microbial communities in a cohort of adult men. Nat Microbiol 2018;3:356–66.10.1038/s41564-017-0084-4Search in Google Scholar PubMed PubMed Central

52. Harnett J, Myers SP, Rolfe M. Significantly higher faecal counts of the yeasts candida and saccharomyces identified in people with coeliac disease. Gut Pathog 2017;9:26.10.1186/s13099-017-0173-1Search in Google Scholar PubMed PubMed Central

53. D’Argenio V, Casaburi G, Precone V, Pagliuca C, Colicchio R, Sarnataro D, et al. No change in the mucosal gut mycobioma is associated with celiac disease-specific microbiome alteration in adult patients. Am J Gastroenterol 2016;111:1659–61.10.1038/ajg.2016.227Search in Google Scholar PubMed

54. Bouziat R, Hinterleitner R, Brown JJ, Stencel-Baerenwald JE, Ikizler M, Mayassi T, et al. Reovirus infection triggers inflammatory responses to dietary antigens and development of celiac disease. Science 2017;356:44–50.10.1126/science.aah5298Search in Google Scholar PubMed PubMed Central

55. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, et al. Enterotypes of the human gut microbiome. Nature 2011;473:174–80.10.1038/nature09944Search in Google Scholar PubMed PubMed Central

56. Flint HJ, Scott KP, Louis P, Duncan SH. The role of the gut microbiota in nutrition and health. Nat Rev Gastroenterol Hepatol 2012;9:577–89.10.1038/nrgastro.2012.156Search in Google Scholar PubMed

57. de Meij TG, Budding AE, Grasman ME, Kneepkens CM, Savelkoul PH, Mearin ML. Composition and diversity of the duodenal mucosa-associated microbiome in children with untreated coeliac disease. Scand J Gastroenterol 2013;48:530–6.10.3109/00365521.2013.775666Search in Google Scholar PubMed

58. Cheng J, Kalliomäki M, Heilig HG, Palva A, Lähteenoja H, de Vos WM, et al. Duodenal microbiota composition and mucosal homeostasis in pediatric celiac disease. BMC Gastroenterol 2013;13:113.10.1186/1471-230X-13-113Search in Google Scholar PubMed PubMed Central

59. Nistal E, Caminero A, Herrán AR, Pérez-Andres J, Vivas S, Ruiz de Morales JM. Study of duodenal bacterial communities by 16S rRNA gene analysis in adults with active celiac disease vs non-celiac disease controls. J Appl Microbiol 2016;120:1691–700.10.1111/jam.13111Search in Google Scholar PubMed

60. Sánchez E, Nadal I, Donat E, Ribes-Koninckx C, Calabuig M, Sanz Y. Reduced diversity and increased virulence-gene carriage in intestinal enterobacteria of coeliac children. BMC Gastroenterol 2008;8:50.10.1186/1471-230X-8-50Search in Google Scholar PubMed PubMed Central

61. Sánchez E, Laparra JM, Sanz Y. Discerning the role of Bacteroides fragilis in celiac disease pathogenesis. Appl Environ Microbiol 2012;78:6507–15.10.1128/AEM.00563-12Search in Google Scholar PubMed PubMed Central

62. Labruna G, Nanayakkara M, Pagliuca C, Nunziato M, Iaffaldano L, D’Argenio V, et al. Celiac disease-associated Neisseria flavescens decreases mitochondrial respiration in CaCo-2 epithelial cells: impact of Lactobacillus paracasei CBA L74 on bacterial-induced cellular imbalance. Cell Microbiol 2019;21:e13035.10.1111/cmi.13035Search in Google Scholar PubMed PubMed Central

63. Helmerhorst EJ, Zamakhchari M, Schuppan D, Oppenheim FG. Discovery of a novel and rich source of gluten-degrading microbial enzymes in the oral cavity. PLoS One 2010;5:e13264.10.1371/journal.pone.0013264Search in Google Scholar PubMed PubMed Central

64. Zamakhchari M, Wei G, Dewhirst F, Lee J, Schuppan D, Oppenheim FG, et al. Identification of Rothia bacteria as gluten-degrading natural colonizers of the upper gastro-intestinal tract. PLoS One 2011;6:e24455.10.1371/journal.pone.0024455Search in Google Scholar PubMed PubMed Central

65. Fernandez-Feo M, Wei G, Blumenkranz G, Dewhirst FE, Schuppan D, Oppenheim FG, et al. The cultivable human oral gluten-degrading microbiome and its potential implications in coeliac disease and gluten sensitivity. Clin Microbiol Infect 2013;19:E386–94.10.1111/1469-0691.12249Search in Google Scholar PubMed PubMed Central

66. Caminero A, McCarville JL, Galipeau HJ, Deraison C, Bernier SP, Constante M, et al. Duodenal bacterial proteolytic activity determines sensitivity to dietary antigen through protease-activated receptor-2. Nat Commun 2019;10:1198.10.1038/s41467-019-09037-9Search in Google Scholar PubMed PubMed Central

67. Stricker S, de Laffolie J, Rudloff S, Komorowski L, Zimmer KP. Intracellular localization of microbial transglutaminase and its influence on the transport of gliadin in enterocytes. J Pediatr Gastroenterol Nutr 2019;68:e43–50.10.1097/MPG.0000000000002171Search in Google Scholar PubMed

68. Torsten M, Aaron L. Microbial transglutaminase is immunogenic and potentially pathogenic in pediatric celiac disease. Front Pediatr 2018;6:389.10.3389/fped.2018.00389Search in Google Scholar PubMed PubMed Central

69. Moerkens R, Mooiweer J, Withoff S, Wijmenga C. Celiac disease-on-chip: modeling a multifactorial disease in vitro. United European Gastroenterol J 2019;7:467–76.10.1177/2050640619836057Search in Google Scholar PubMed PubMed Central

70. Kasendra M, Tovaglieri A, Sontheimer-Phelps A, Jalili-Firoozinezhad S, Bein A, Chalkiadaki A, et al. Development of a primary human Small Intestine-on-a-Chip using biopsy-derived organoids. Sci Rep 2018;8:2871.10.1038/s41598-018-21201-7Search in Google Scholar PubMed PubMed Central


Supplementary Material

The online version of this article offers supplementary material (https://doi.org/10.1515/cclm-2019-0657).


Received: 2019-06-28
Accepted: 2019-08-06
Published Online: 2019-09-09
Published in Print: 2020-02-25

©2019 Lucia Sacchetti et al., published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

Articles in the same Issue

  1. Frontmatter
  2. Editorial
  3. Progress in understanding the use of human chorionic gonadotropin as a tumor marker
  4. Reviews
  5. Biomarkers for prostate cancer: prostate-specific antigen and beyond
  6. Gut microbiome investigation in celiac disease: from methods to its pathogenetic role
  7. Opinion Papers
  8. Understanding and managing interferences in clinical laboratory assays: the role of laboratory professionals
  9. A step forward in identifying the right human chorionic gonadotropin assay for testicular cancer
  10. EFLM Opinion Paper
  11. Validation and verification of examination procedures in medical laboratories: opinion of the EFLM Working Group Accreditation and ISO/CEN standards (WG-A/ISO) on dealing with ISO 15189:2012 demands for method verification and validation
  12. Guidelines and Recommendations
  13. An updated protocol based on CLSI document C37 for preparation of off-the-clot serum from individual units for use alone or to prepare commutable pooled serum reference materials
  14. General Clinical Chemistry and Laboratory Medicine
  15. Highly accurate and explainable detection of specimen mix-up using a machine learning model
  16. Impact of delta check time intervals on error detection capability
  17. Effect of long-term frozen storage and thawing of stool samples on faecal haemoglobin concentration and diagnostic performance of faecal immunochemical tests
  18. A more accurate prediction to rule in and rule out pre-eclampsia using the sFlt-1/PlGF ratio and NT-proBNP as biomarkers
  19. Macro vitamin B12: an underestimated threat
  20. Evaluation of the primary biliary cholangitis-related serologic profile in a large cohort of Belgian systemic sclerosis patients
  21. High frequency of anti-parietal cell antibody (APCA) and intrinsic factor blocking antibody (IFBA) in individuals with severe vitamin B12 deficiency – an observational study in primary care patients
  22. DOAC-Remove abolishes the effect of direct oral anticoagulants on activated protein C resistance testing in real-life venous thromboembolism patients
  23. Cancer Diagnostics
  24. How comparable are total human chorionic gonadotropin (hCGt) tumour markers assays?
  25. Diabetes
  26. Measurement accuracy of two professional-use systems for point-of-care testing of blood glucose
  27. Letters to the Editor
  28. Human chorionic gonadotropin in oncology: a matter of tight (bio)marking
  29. More robust analytical evidence should support the selection of human chorionic gonadotropin assays for oncology application
  30. Letter in reply to the letter to the editor of Ferraro S and Panteghini M with the title “More robust analytical evidence should support the selection for human chorionic gonadotropin assays for oncology application”
  31. Definition of analytical quality specifications for serum total folate measurements using a simulation outcome-based model
  32. Alarmed by misleading interference in free T3 and free T4 assays: a new case of anti-streptavidin antibodies
  33. A root cause analysis of ‘falsely elevated’ oxygen saturation: investigation of pneumatic tube transport and differences between estimated and measured saturation in a critical patient population
  34. The chaos of serologic markers in interstitial pneumonia with autoimmune features can be corrected by the laboratory physician
  35. Vitamin A and E gender and age stratification in adults
  36. Detection of monoclonal B-lymphocytosis: interest of cellular population data and CytoDiff™ analysis
  37. Urinary reference intervals for gadolinium in individuals without recent exposure to gadolinium-based contrast agents
  38. Molecular diagnosis of toxoplasmosis: evaluation of automated DNA extraction using eMAG® (bioMérieux) on buffy coat, cerebrospinal and bronchoalveolar lavage fluids
  39. Retraction
  40. Retraction of: The Effect of a Gender Difference in the Apolipoprotein E Gene DNA Polymorphism on Serum Lipid Levels in a Serbian Healthy Population
Downloaded on 22.12.2025 from https://www.degruyterbrill.com/document/doi/10.1515/cclm-2019-0657/html
Scroll to top button