Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Biol Sex Differ ; 14(1): 86, 2023 12 11.
Article En | MEDLINE | ID: mdl-38072919

BACKGROUND: Celiac disease (CeD) is an immune-mediated disorder that develops in genetically predisposed individuals upon gluten consumption. HLA risk alleles explain 40% of the genetic component of CeD, so there have been continuing efforts to uncover non-HLA loci that can explain the remaining heritability. As in most autoimmune disorders, the prevalence of CeD is significantly higher in women. Here, we investigated the possible involvement of the X chromosome on the sex bias of CeD. METHODS: We performed a X chromosome-wide association study (XWAS) and a gene-based association study in women from the CeD Immunochip (7062 cases, 5446 controls). We also constructed a database of X chromosome cis-expression quantitative trait loci (eQTLs) in monocytes from unstimulated (n = 226) and lipopolysaccharide (LPS)-stimulated (n = 130) female donors and performed a Summary-data-based MR (SMR) analysis to integrate XWAS and eQTL information. We interrogated the expression of the potentially causal gene (TMEM187) in peripheral blood mononuclear cells (PBMCs) from celiac patients at onset, on a gluten-free diet, potential celiac patients and non-celiac controls. RESULTS: The XWAS and gene-based analyses identified 13 SNPs and 25 genes, respectively, 22 of which had not been previously associated with CeD. The X chromosome cis-eQTL analysis found 18 genes with at least one cis-eQTL in naïve female monocytes and 8 genes in LPS-stimulated female monocytes, 2 of which were common to both situations and 6 were unique to LPS stimulation. SMR identified a potentially causal association of TMEM187 expression in naïve monocytes with CeD in women, regulated by CeD-associated, eQTL-SNPs rs7350355 and rs5945386. The CeD-risk alleles were correlated with lower TMEM187 expression. These results were replicated using eQTLs from LPS-stimulated monocytes. We observed higher levels of TMEM187 expression in PBMCs from female CeD patients at onset compared to female non-celiac controls, but not in male CeD individuals. CONCLUSION: Using X chromosome genotypes and gene expression data from female monocytes, SMR has identified TMEM187 as a potentially causal candidate in CeD. Further studies are needed to understand the implication of the X chromosome in the higher prevalence of CeD in women.


Celiac disease (CeD) is an immune-related condition triggered by gluten consumption in genetically susceptible individuals. Women present higher prevalence of CeD than men, but the biological explanation of such difference has not been elucidated. In this study, we investigated whether specific genetic variations on the X chromosome were associated with CeD in each sex. Surprisingly, we found 13 genetic variants and 25 genes significantly linked to CeD in women, but not in men. Additionally, we identified genetic variants on the X chromosome associated with gene expression of monocytes, a type of immune cells that is activated in CeD after gluten intake. Integrating these data with our previous findings, we found that lower expression of a gene termed TMEM187 might be associated with a potential increase in CeD risk in women. Finally, validation experiments confirmed higher TMEM187 levels in blood cells from female CeD patients compared to non-celiac women, while no such difference was seen in males. In summary, our study suggests that the X-chromosome gene TMEM187 may play a key role in CeD development, providing insights into the higher prevalence of CeD in females.


Celiac Disease , Quantitative Trait Loci , Humans , Male , Female , Celiac Disease/genetics , Celiac Disease/metabolism , Monocytes/metabolism , Leukocytes, Mononuclear , Sexism , Lipopolysaccharides , Membrane Proteins/genetics
2.
Gut ; 71(1): 68-76, 2022 01.
Article En | MEDLINE | ID: mdl-33526437

OBJECTIVES: Coeliac disease (CD) is a complex autoimmune disorder that develops in genetically susceptible individuals. Dietary gluten triggers an immune response for which the only available treatment so far is a strict, lifelong gluten free diet. Human leucocyte antigen (HLA) genes and several non-HLA regions have been associated with the genetic susceptibility to CD, but their role in the pathogenesis of the disease is still essentially unknown, making it complicated to develop much needed non-dietary treatments. Here, we describe the functional involvement of a CD-associated single-nucleotide polymorphism (SNP) located in the 5'UTR of XPO1 in the inflammatory environment characteristic of the coeliac intestinal epithelium. DESIGN: The function of the CD-associated SNP was investigated using an intestinal cell line heterozygous for the SNP, N6-methyladenosine (m6A)-related knock-out and HLA-DQ2 mice, and human samples from patients with CD. RESULTS: Individuals harbouring the risk allele had higher m6A methylation in the 5'UTR of XPO1 RNA, rendering greater XPO1 protein amounts that led to downstream nuclear factor kappa B (NFkB) activity and subsequent inflammation. Furthermore, gluten exposure increased overall m6A methylation in humans as well as in in vitro and in vivo models. CONCLUSION: We identify a novel m6A-XPO1-NFkB pathway that is activated in CD patients. The findings will prompt the development of new therapeutic approaches directed at m6A proteins and XPO1, a target under evaluation for the treatment of intestinal disorders.


Celiac Disease/genetics , Karyopherins/genetics , Polymorphism, Single Nucleotide , RNA/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Adenosine/analogs & derivatives , Adenosine/genetics , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/pathology , HLA-DQ Antigens/genetics , Humans , Intestinal Mucosa/pathology , Methylation , Mice, Knockout , NF-kappa B/metabolism , Exportin 1 Protein
3.
Front Immunol ; 10: 2325, 2019.
Article En | MEDLINE | ID: mdl-31681265

Primary immunodeficiencies (PIDs) refer to a clinically, immunologically, and genetically heterogeneous group of over 350 disorders affecting development or function of the immune system. The increasing use of next-generation sequencing (NGS) technology has greatly facilitated identification of genetic defects in PID patients in daily clinical practice. Several NGS approaches are available, from the unbiased whole exome sequencing (WES) to specific gene panels. Here, we report on a 3-year experience with clinical exome sequencing (CES) for genetic diagnosis of PIDs. We used the TruSight One sequencing panel, which includes 4,813 disease-associated genes, in 61 unrelated patients (pediatric and adults). The analysis was done in 2 steps: first, we focused on a virtual PID panel and then, we expanded the analysis to the remaining genes. A molecular diagnosis was achieved in 19 (31%) patients: 12 (20%) with mutations in genes included in the virtual PID panel and 7 (11%) with mutations in other genes. These latter cases provided interesting and somewhat unexpected findings that expand the clinical and genetic spectra of PID-related disorders, and are useful to consider in the differential diagnosis. We also discuss 5 patients (8%) with incomplete genotypes or variants of uncertain significance. Finally, we address the limitations of CES exemplified by 7 patients (11%) with negative results on CES who were later diagnosed by other approaches (more specific PID panels, WES, and comparative genomic hybridization array). In summary, the genetic diagnosis rate using CES was 31% (including a description of 12 novel mutations), which rose to 42% after including diagnoses achieved by later use of other techniques. The description of patients with mutations in genes not included in the PID classification illustrates the heterogeneity and complexity of PID-related disorders.


Genetic Association Studies , Genetic Predisposition to Disease , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Adolescent , Adult , Child , Child, Preschool , Female , Genetic Association Studies/methods , Genetic Variation , Genotype , Humans , Infant , Infant, Newborn , Male , Middle Aged , Mutation , Phenotype , Primary Immunodeficiency Diseases/diagnosis , Exome Sequencing , Young Adult
4.
Sci Rep ; 9(1): 1298, 2019 02 04.
Article En | MEDLINE | ID: mdl-30718669

The Human Leucocyte Antigen (HLA) locus and other DNA sequence variants identified in Genome-Wide Association (GWA) studies explain around 50% of the heritability of celiac disease (CD). However, the pathogenesis of CD could be driven by other layers of genomic information independent from sequence variation, such as DNA methylation, and it is possible that allele-specific methylation explains part of the SNP associations. Since the DNA methylation landscape is expected to be different among cell types, we analyzed the methylome of the epithelial and immune cell populations of duodenal biopsies in CD patients and controls separately. We found a cell type-specific methylation signature that includes genes mapping to the HLA region, namely TAP1 and HLA-B. We also performed Immunochip SNP genotyping of the same samples and interrogated the expression of some of the affected genes. Our analysis revealed that the epithelial methylome is characterized by the loss of CpG island (CGI) boundaries, often associated to altered gene expression, and by the increased variability of the methylation across the samples. The overlap between differentially methylated positions (DMPs) and CD-associated SNPs or variants contributing to methylation quantitative trait loci (mQTLs) is minimal. In contrast, there is a notable enrichment of mQTLs among the most significant CD-associated SNPs. Our results support the notion that DNA methylation alterations constitute a genotype-independent event and confirm its role in the HLA region (apart from the well-known, DQ allele-specific effect). Finally, we find that a fraction of the CD-associated variants could exert its phenotypic effect through DNA methylation.


Celiac Disease/etiology , DNA Methylation , Epigenome , Genotype , HLA Antigens/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Biopsy , Celiac Disease/metabolism , Celiac Disease/pathology , CpG Islands , Female , Gene Expression Profiling , Gene Expression Regulation , Genome-Wide Association Study , HLA Antigens/immunology , Humans , Intestinal Mucosa/pathology , Male , Promoter Regions, Genetic
5.
Front Nutr ; 6: 187, 2019.
Article En | MEDLINE | ID: mdl-31921880

Celiac disease (CD) patients present a loss of intestinal barrier function due to structural alterations in the tight junction (TJ) network, the most apical unions between epithelial cells. The association of TJ-related gene variants points to an implication of this network in disease susceptibility. This work aims to characterize the functional implication of TJ-related, disease-associated loci in CD pathogenesis. We performed an association study of 8 TJ-related gene variants in a cohort of 270 CD and 91 non-CD controls. The expression level of transcripts located in the associated SNP region was analyzed by RT-PCR in several human tissues and in duodenal biopsies of celiac patients and non-CD controls. (si)RNA-driven silencing combined with gliadin in the Caco2 intestinal cell line was used to analyze the implication of transcripts from the associated region in the regulation of TJ genes. We replicated the association of rs6962966*A variant [p = 0.0029; OR = 1.88 (95%1.24-2.87)], located in an intron of TJ-related MAGI2 coding gene and upstream of RP4-587D13.2 transcript, bioinformatically classified as a long non-coding RNA (lncRNA). The expression of both genes is correlated and constitutively downregulated in CD intestine. Silencing of lncRNA decreases the levels of MAGI2 protein. At the same time, silencing of MAGI2 affects the expression of several TJ-related genes. The associated region is functionally altered in disease, probably affecting CD-related TJ genes.

6.
Genes (Basel) ; 9(5)2018 May 10.
Article En | MEDLINE | ID: mdl-29748492

The aim of this study was to construct celiac co-expression patterns at a whole genome level and to identify transcription factors (TFs) that could drive the gliadin-related changes in coordination of gene expression observed in celiac disease (CD). Differential co-expression modules were identified in the acute and chronic responses to gliadin using expression data from a previous microarray study in duodenal biopsies. Transcription factor binding site (TFBS) and Gene Ontology (GO) annotation enrichment analyses were performed in differentially co-expressed genes (DCGs) and selection of candidate regulators was performed. Expression of candidates was measured in clinical samples and the activation of the TFs was further characterized in C2BBe1 cells upon gliadin challenge. Enrichment analyses of the DCGs identified 10 TFs and five were selected for further investigation. Expression changes related to active CD were detected in four TFs, as well as in several of their in silico predicted targets. The activation of TFs was further characterized in C2BBe1 cells upon gliadin challenge, and an increase in nuclear translocation of CAMP Responsive Element Binding Protein 1 (CREB1) and IFN regulatory factor-1 (IRF1) in response to gliadin was observed. Using transcriptome-wide co-expression analyses we are able to propose novel genes involved in CD pathogenesis that respond upon gliadin stimulation, also in non-celiac models.

7.
J Pediatr Gastroenterol Nutr ; 67(2): 225-231, 2018 08.
Article En | MEDLINE | ID: mdl-29601440

OBJECTIVE: The aim of the study is to identify additional celiac disease associated loci in the major histocompatibility complex (MHC) independent from classical HLA risk alleles (HLA-DR3-DQ2) and to characterize their potential functional impact in celiac disease pathogenesis at the intestinal level. METHODS: We performed a high-resolution single-nucleotide polymorphism (SNP) genotyping of the MHC region, comparing HLA-DR3 homozygous celiac patients and non-celiac controls carrying a single copy of the B8-DR3-DQ2 conserved extended haplotype. Expression level of potential novel risk genes was determined by RT-PCR in intestinal biopsies and in intestinal and immune cells isolated from control and celiac individuals. Small interfering RNA-driven silencing of selected genes was performed in the intestinal cell line T84. RESULTS: MHC genotyping revealed 2 associated SNPs, one located in TRIM27 gene and another in the non-coding gene HCG14. After stratification analysis, only HCG14 showed significant association independent from HLA-DR-DQ loci. Expression of HCG14 was slightly downregulated in epithelial cells isolated from duodenal biopsies of celiac patients, and eQTL analysis revealed that polymorphisms in HCG14 region were associated with decreased NOD1 expression in duodenal intestinal cells. CONCLUSIONS: We have successfully employed a conserved extended haplotype-matching strategy and identified a novel additional celiac disease risk variant in the lncRNA HCG14. This lncRNA seems to regulate the expression of NOD1 in an allele-specific manner. Further functional studies are needed to clarify the role of HCG14 in the regulation of gene expression and to determine the molecular mechanisms by which the risk variant in HCG14 contributes to celiac disease pathogenesis.


Celiac Disease/genetics , Genetic Predisposition to Disease , HLA-DR3 Antigen/genetics , Nod1 Signaling Adaptor Protein/metabolism , RNA, Long Noncoding/genetics , Case-Control Studies , Celiac Disease/metabolism , Celiac Disease/pathology , Child , Female , Humans , Male , Polymerase Chain Reaction , Polymorphism, Single Nucleotide
8.
J Immunol ; 199(2): 581-588, 2017 07 15.
Article En | MEDLINE | ID: mdl-28626066

Long noncoding RNAs (lncRNAs) have emerged as critical regulators of inflammation. To further understand the interaction between inflammatory signaling pathways and lncRNAs, we characterized the function of cardiac and apoptosis-related lncRNA (Carlr), an lncRNA expressed in both mouse and human cells of diverse tissues. Carlr expression is increased following NF-κB signaling in macrophages, with concomitant translocation to, and enrichment of, the transcript in the cytoplasm. Knockdown of Carlr results in impaired expression of NF-κB pathway genes and influences the interaction between macrophages and intestinal cells in an inflammatory environment. In human celiac disease patient samples, increased levels of the Carlr transcript were detected in the cytoplasm, alongside elevated expression of NF-κB pathway genes. These findings suggest that increased Carlr expression and/or cytoplasmic localization is required for efficient NF-κB signaling and is associated with the inflamed tissue state observed in human celiac disease.


Cytoplasm/genetics , Gene Expression Regulation , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , Animals , Apoptosis , Celiac Disease/immunology , Celiac Disease/metabolism , Cytoplasm/immunology , Cytoplasm/metabolism , Gene Expression , Humans , Inflammation , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , NF-kappa B/immunology , Phosphorylation , Signal Transduction
9.
Eur J Hum Genet ; 24(12): 1831-1834, 2016 12.
Article En | MEDLINE | ID: mdl-27650971

To identify candidate genes in celiac disease (CD), we reanalyzed the whole Immunochip CD cohort using a different approach that clusters individuals based on immunoancestry prior to disease association analysis, rather than by geographical origin. We detected 636 new associated SNPs (P<7.02 × 10-07) and identified 5 novel genomic regions, extended 8 others previously identified and also detected 18 isolated signals defined by one or very few significant SNPs. To test whether we could identify putative candidate genes, we performed expression analyses of several genes from the top novel region (chr2:134533564-136169524), from a previously identified locus that is now extended, and a gene marked by an isolated SNP, in duodenum biopsies of active and treated CD patients, and non-celiac controls. In the largest novel region, CCNT2 and R3HDM1 were constitutively underexpressed in disease, even after gluten removal. Moreover, several genes within this region were coexpressed in patients, but not in controls. Other novel genes like KIF21B, REL and SORD also showed altered expression in active disease. Apart from the identification of novel CD loci, these results suggest that ancestry-based stratified analysis is an efficient strategy for association studies in complex diseases.


Celiac Disease/genetics , Genetic Loci , Pedigree , Polymorphism, Single Nucleotide , Case-Control Studies , Celiac Disease/metabolism , Duodenum/metabolism , Glutens/metabolism , Humans
10.
Clin Nutr ; 35(6): 1406-1413, 2016 12.
Article En | MEDLINE | ID: mdl-27086197

BACKGROUND & AIMS: Pulmonary embolism (PE) is a complication of parenteral nutrition (PN) with a prevalence of 35% in children. In 2003 new intravenous lipid emulsions (ILEs) with MCT, olive and/or fish oil in addition to soybean oil were introduced. The aim was to compare the incidence of PE before and after introduction. METHODS: 327 surveillance ventilation-perfusion (V/Q) scintigraphies from 68 children aged 0.3-15 years, treated with PN from 1993 to 2010, were retrospectively reviewed. Rate of PE/1000 central venous catheter (CVC) days, number of children with PE pre- and post-introduction of ILEs were compared. Multivariate analyses were performed for risk factors. RESULTS: Twenty-two (32%) children (19/42 before 2003 and 3/26 after 2003, p = 0.007) had at least one episode of PE. Thirty seven (11%) episodes of PE were detected accounting for a mean of 0.2/1000 CVC days prior to 2003 and 0.05/1000 CVC days after 2003, p = 0.04. Regression analysis indicated that higher content of ILE/infusion (p = 0.045) and frequency of ILE of >3 nights/week were associated with more PE (p = 0.001). New ILEs were associated with lower risk (p = 0.003). CONCLUSION: With a four-fold fall in incidence with new ILE, PE remains a complication. We recommend 12-18 monthly surveillance with lung perfusion scan and anticoagulants if PE is diagnosed.


Parenteral Nutrition/adverse effects , Pulmonary Embolism/epidemiology , Pulmonary Embolism/prevention & control , Adolescent , Central Venous Catheters , Child , Child, Preschool , Fat Emulsions, Intravenous/administration & dosage , Female , Fish Oils/administration & dosage , Follow-Up Studies , Humans , Incidence , Infant , Male , Olive Oil/administration & dosage , Retrospective Studies , Risk Factors , Soybean Oil/administration & dosage , Triglycerides/administration & dosage
11.
Eur J Hum Genet ; 23(8): 1100-5, 2015 Aug.
Article En | MEDLINE | ID: mdl-25388004

Celiac disease is a chronic immune-mediated disorder with an important genetic component. To date, there are 57 independent association signals from 39 non-HLA loci, and a total of 66 candidate genes have been proposed. We aimed to scrutinize the functional implication of 45 of those genes by analyzing their expression in the disease tissue of celiac patients (at diagnosis/treatment) compared with non-celiac controls. Moreover, we investigated the SNP genotype effect in gene expression and performed coexpression analyses. Several genes showed differential expression among disease groups, most of them related to immune response. Multiple trans-eQTLs but only four cis-eQTLs were found, and surprisingly the genotype effect seems to be stimulus dependent as it differs among groups. Coexpression levels vary from higher to lower levels in active patients at diagnosis, treated patients and non-celiac controls respectively. A subset of 18 genes tightly correlated in both groups of patients but not in controls was identified. Interestingly, this subset of genes was influenced by the genotype of three SNPs. One of the SNPs, rs1018326 on chromosome two is on top of a known lincRNA whose function is not yet described, and whose expression seems to be upregulated in active disease when comparing biopsy pairs from the same individuals. Our results strongly suggest that the effects of disease-associated SNPs go far beyond the oversimplistic idea of transcriptional control at a nearby locus. Further investigations are needed to determine how each variant disrupts fine-tuning mechanisms in the genome that eventually lead to disease.


Celiac Disease/genetics , Genetic Predisposition to Disease , Intestinal Mucosa/metabolism , Quantitative Trait Loci/genetics , Celiac Disease/metabolism , Celiac Disease/pathology , Female , Gene Expression Regulation , Genetic Association Studies , Humans , Intestinal Mucosa/pathology , Male , Polymorphism, Single Nucleotide , RNA, Long Noncoding/genetics
12.
World J Gastroenterol ; 20(5): 1357-60, 2014 Feb 07.
Article En | MEDLINE | ID: mdl-24574811

Celiac disease is a chronic, immune-mediated enteropathy caused by a permanent sensitivity to ingested gluten cereals that develops in genetically susceptible individuals. The classic presentation of celiac disease includes symptoms of malabsorption but has long been associated with cognitive, emotional, and behavioral disorders. We describe an 8-year-old patient with non-scarring alopecia and diagnosed with trichotillomania. Furthermore, she presented with a 3-year history of poor appetite and two or three annual episodes of mushy, fatty stools. Laboratory investigations showed a normal hemoglobin concentration and a low ferritin level. Serologic studies showed an elevated tissue immunoglobulin G anti-tissue transglutaminase level. A duodenal biopsy showed subtotal villous atrophy and crypt hyperplasia, and a large gastric trichobezoar was found in the stomach. Immediately after beginning a gluten-free diet, complete relief of trichotillomania and trichophagia was achieved. In this report, we describe a behavioral disorder as a primary phenomenon of celiac disease, irrespective of nutritional status.


Bezoars/etiology , Celiac Disease/diagnosis , Stomach , Trichotillomania/etiology , Bezoars/diagnosis , Celiac Disease/diet therapy , Celiac Disease/psychology , Child , Child Behavior , Diet, Gluten-Free , Emotions , Female , Humans , Treatment Outcome , Trichotillomania/diagnosis , Trichotillomania/psychology
13.
BMC Gastroenterol ; 14: 24, 2014 Feb 11.
Article En | MEDLINE | ID: mdl-24517104

BACKGROUND: The World Gastroenterology Organization recommends developing national guidelines for the diagnosis of Celiac Disease (CD): hence a profile of the diagnosis of CD in each country is required. We aim to describe a cross-sectional picture of the clinical features and diagnostic facilities in 16 countries of the Mediterranean basin. Since a new ESPGHAN diagnostic protocol was recently published, our secondary aim is to estimate how many cases in the same area could be identified without a small intestinal biopsy. METHODS: By a stratified cross-sectional retrospective study design, we examined clinical, histological and laboratory data from 749 consecutive unselected CD children diagnosed by national referral centers. RESULTS: The vast majority of cases were diagnosed before the age of 10 (median: 5 years), affected by diarrhea, weight loss and food refusal, as expected. Only 59 cases (7.8%) did not suffer of major complaints. Tissue transglutaminase (tTG) assay was available, but one-third of centers reported financial constraints in the regular purchase of the assay kits. 252 cases (33.6%) showed tTG values over 10 times the local normal limit. Endomysial antibodies and HLA typing were routinely available in only half of the centers. CD was mainly diagnosed from small intestinal biopsy, available in all centers. Based on these data, only 154/749 cases (20.5%) would have qualified for a diagnosis of CD without a small intestinal biopsy, according to the new ESPGHAN protocol. CONCLUSIONS: This cross-sectional study of CD in the Mediterranean referral centers offers a puzzling picture of the capacities to deal with the emerging epidemic of CD in the area, giving a substantive support to the World Gastroenterology Organization guidelines.


Biopsy/statistics & numerical data , Celiac Disease/diagnosis , Genotyping Techniques/statistics & numerical data , Intestine, Small/pathology , Serologic Tests/statistics & numerical data , Adolescent , Africa, Northern , Anorexia/etiology , Antibodies/blood , Celiac Disease/genetics , Celiac Disease/pathology , Child , Child, Preschool , Cross-Sectional Studies , Diarrhea/etiology , Europe, Eastern , Female , GTP-Binding Proteins , HLA Antigens/genetics , Haplotypes , Humans , Infant , Male , Mediterranean Region , Practice Guidelines as Topic , Protein Glutamine gamma Glutamyltransferase 2 , Retrospective Studies , Transglutaminases/blood , Vomiting/etiology , Weight Loss
14.
J Pediatr Gastroenterol Nutr ; 58(6): 762-7, 2014 Jun.
Article En | MEDLINE | ID: mdl-24552675

OBJECTIVE: The aim of the present study was to characterize the deregulation of epithelial tight junction genes and investigate its reversibility on removal of dietary gluten in small intestinal mucosa in celiac disease (CD). METHODS: The expression levels of 23 genes related to tight junctions were studied in biopsies from 16 patients with active CD and compared with biopsies from the same patients taken after 2 years on gluten-free diet (GFD) and with 16 non-CD controls. RESULTS: Nine genes showed altered expression levels in patients with active disease (CLDN2, PARD6A, ZAK, SYMPK, MYH14, and ACTB were upregulated, whereas MAGI1, TJP1, and PPP2R3A were downregulated). Alterations were reversible after 2 years on treatment, except for PPP2R3A, implicated in the negative control of cell growth and division. At the biological network level, important dysfunctions in several processes within the pathway were observed, including intestinal permeability, apicobasal polarity, and cell proliferation. CONCLUSIONS: Our work confirms the involvement of tight junction genes related to permeability, polarity, and cell proliferation in the epithelial destruction observed in CD. Coexpression patterns of several genes support the idea of a common regulatory mechanism that seems to be altered in active CD. In general, GFD normalization confirms the reversibility of the process, except for the constitutive downregulation of PPP2R3A suggestive of a genetic implication. Further studies in proteins and cells or tissues are necessary to confirm these findings.


Celiac Disease/genetics , Gene Expression , Intestinal Mucosa/pathology , Intestine, Small/pathology , Tight Junctions , Celiac Disease/diet therapy , Cell Polarity , Cell Proliferation , Child , Child, Preschool , Diet, Gluten-Free , Down-Regulation , Humans , Infant , Male , Permeability , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism
15.
Hum Mol Genet ; 23(5): 1298-310, 2014 Mar 01.
Article En | MEDLINE | ID: mdl-24163129

It is known that the NFκB route is constitutively upregulated in celiac disease (CD), an immune-mediated disorder of the gut caused by intolerance to ingested gluten. Our aim was to scrutinize the expression patterns of several of the most biologically relevant components of the NFκB route in intestinal biopsies from active and treated patients and after in vitro gliadin challenge, and to assess normalization of the expression using an inhibitor of the MALT1 paracaspase. The expression of 93 NFκB genes was measured by RT-PCR in a set of uncultured active and treated CD and control biopsies, and in cultured biopsy series challenged with gliadin, the NFκB modulator, both compounds and none. Methylation of eight genes involved in NFκB signaling was analyzed by conventional pyrosequencing. Groups were compared and Pearson's correlation matrixes were constructed to check for coexpression and co-methylation. Our results confirm the upregulation of the NFκB pathway and show that constitutively altered genes usually belong to the core of the pathway and have central roles, whereas genes overexpressed only in active CD are more peripheral. Additionally, this is the first work to detect methylation level changes in celiac intestinal mucosa. Coexpression is very common in controls, whereas gliadin challenge and especially chronic inflammation present in untreated CD result in the disruption of the regulatory equilibrium. In contrast, co-methylation occurs more often in active CD. Importantly, NFκB modulation partially restores coregulation, opening the door to future therapeutic possibilities and targets.


Celiac Disease/genetics , Celiac Disease/metabolism , Gene Expression Regulation , NF-kappa B/metabolism , Cluster Analysis , DNA Methylation , Gene Expression , Gene Expression Profiling , Gene Regulatory Networks , Humans , Inflammation/genetics , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Promoter Regions, Genetic , Signal Transduction
16.
Eur J Hum Genet ; 22(3): 358-62, 2014 Mar.
Article En | MEDLINE | ID: mdl-23820479

Celiac disease (CD) is an immune mediated, polygenic disorder, where HLA-DQ2/DQ8 alleles contribute around 35% to genetic risk, but several other genes are also involved. Genome-wide association studies (GWASs) and the more recent immunochip genotyping projects have fine-mapped 39 regions of genetic susceptibility to the disease, most of which harbor candidate genes that could participate in this disease process. We focused our attention to the GWAS peak on chr6: 127.99-128.38 Mb, a region including two genes, thymocyte-expressed molecule involved in selection (THEMIS) and protein tyrosine phosphatase, receptor type, kappa (PTPRK), both of which have immune-related functions. The aim of this work was to evaluate the expression levels of these two genes in duodenal mucosa of active and treated CD patients and in controls, and to determine whether SNPs (rs802734, rs55743914, rs72975916, rs10484718 and rs9491896) associated with CD have any influence on gene expression. THEMIS showed higher expression in active CD compared with treated patients and controls, whereas PTPRK showed lower expression. Our study confirmed the association of this region with CD in our population, but only the genotype of rs802734 showed some influence in the expression of THEMIS. On the other hand, we found a significant positive correlation between THEMIS and PTPRK mRNA levels in CD patients but not in controls. Our results suggest a possible role for both candidate genes in CD pathogenesis and the existence of complex, regulatory relationships that reside in the vast non-coding, functional intergenic regions of the genome. Further investigation is needed to clarify the impact of the disease-associated SNPs on gene function.


Celiac Disease/genetics , Gliadin/pharmacology , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Case-Control Studies , Celiac Disease/metabolism , Cells, Cultured , Child, Preschool , Chromosomes, Human, Pair 6/genetics , Duodenum/metabolism , Duodenum/pathology , Female , Humans , Infant , Intestinal Mucosa/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Male , Polymorphism, Single Nucleotide , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism
17.
Autoimmunity ; 45(3): 264-70, 2012 May.
Article En | MEDLINE | ID: mdl-22136669

Celiac disease (CD) involves disturbance of the small-bowel mucosal vascular network, and transglutaminase autoantibodies (TGA) have been related to angiogenesis disturbance, a complex phenomenon probably also influenced by common genetic variants in angiogenesis-related genes. A set of genes with "angiogenesis" GO term identified in a previous expression microarray experiment (SCG2, STAB1, TGFA, ANG, ERBB2, GNA13, PML, CASP8, ECGF1, JAG1, HIF1A, TNFSF13 and TGM2) was selected for genetic and functional studies. SNPs that showed a trend for association with CD in the first GWAS were genotyped in 555 patients and 541 controls. Gene expression of all genes was quantified in 15 pairs of intestinal biopsies (diagnosis vs. GFD) and in three-dimensional HUVEC and T84 cell cultures incubated with TGA-positive and negative serum. A regulatory SNP in TNFSF13 (rs11552708) is associated with CD (p = 0.01, OR = 0.7). Expression changes in biopsies pointed to TGM2 and PML as up-regulated antiangiogenic genes and to GNA13, TGFA, ERBB2 and SCG2 as down-regulated proangiogenic factors in CD. TGA seem to enhance TGM2 expression in both cell models, but PML expression was induced only in T84 enterocytes while GNA13 and ERBB2 were repressed in HUVEC endothelial cells, with several genes showing discordant effects in each model, highlighting the complexity of gene interactions in the pathogenesis of CD. Finally, cell culture models are useful tools to help dissect complex responses observed in human explants.


Celiac Disease/genetics , Gene Expression Profiling , Neovascularization, Pathologic/genetics , Biopsy , Celiac Disease/pathology , Cell Line , Cells, Cultured , Child, Preschool , Female , Gene Expression Regulation , Genome-Wide Association Study , Humans , Infant , Male , Models, Statistical , Polymorphism, Single Nucleotide
18.
PLoS One ; 6(12): e28910, 2011.
Article En | MEDLINE | ID: mdl-22174923

The possible implication of copy number variation (CNV) in the genetic susceptibility to human disease needs to be assessed using robust methods that can be applied at a population scale. In this report, we analyze the performance of the two major techniques, quantitative PCR (qPCR) and paralog ratio test (PRT), and investigate the influence of input DNA amount and template integrity on the reliability of both methods. Analysis of three genes (PRELID1, SYNPO and DEFB4) in a large sample set showed that both methods are prone to false copy number assignments if sufficient attention is not paid to DNA concentration and quality. Accurate normalization of samples is essential for reproducible qPCR because it avoids the effect of differential amplification efficiencies between target and control assays, whereas PRT is generally more sensitive to template degradation due to the fact that longer amplicons are usually needed to optimize sensitivity and specificity of paralog sequence PCR. The use of normalized, high quality genomic DNA yields comparable results with both methods.


Biological Assay/methods , DNA Copy Number Variations/genetics , DNA/genetics , Polymerase Chain Reaction/methods , Sequence Homology, Nucleic Acid , Humans , Microfilament Proteins/genetics , Mitochondrial Proteins/genetics , Reproducibility of Results , beta-Defensins/genetics
19.
Hum Immunol ; 72(8): 617-20, 2011 Aug.
Article En | MEDLINE | ID: mdl-21616111

Killer cell immunoglobulin-like receptors (KIRs) modulate natural killer (NK) and T-cell function by human leukocyte antigen class I interaction and have been implicated in celiac disease (CD). Qualitative expression of 16 KIR genes was determined in biopsies from 22 CD patients at diagnosis and after >2 years on a gluten-free diet (GFD). Quantitative expression analysis of KIR2DL4, KIR3DL1, KIR3DL3, and KLRC2 (a marker of an NK-reprogrammed T-cell subpopulation augmented in CD) was performed in 35 additional CD biopsy pairs and 14 non-CD control biopsies. No specific KIR expression profile was observed in CD. KIR3DL1 was more frequently expressed in active CD compared with GFD (p = 0.0312) and controls (p = 0.0008), with slightly increased levels in active disease. KLRC2 was overexpressed in active (p = 0.0037) and GFD (p = 0.0469) patients compared with non-CD controls and coexpressed with KIR3DL1. Results suggest the participation of KIR3DL1 overexpression in the overall immune activation seen in CD mucosa, which could be partly explained by the NK-like T-cell subpopulation increase.


Celiac Disease , Intestinal Mucosa/metabolism , Killer Cells, Natural/metabolism , Receptors, KIR2DL1/metabolism , Biopsy , Celiac Disease/diagnosis , Celiac Disease/immunology , Celiac Disease/metabolism , Celiac Disease/pathology , Child, Preschool , Diet, Gluten-Free , Female , Gene Expression , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Killer Cells, Natural/immunology , Male , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Receptors, KIR2DL1/genetics , Receptors, KIR2DL3/genetics , Receptors, KIR2DL3/metabolism , Receptors, KIR2DL4/genetics , Receptors, KIR2DL4/metabolism , Reverse Transcriptase Polymerase Chain Reaction
20.
J Med Genet ; 48(7): 493-6, 2011 Jul.
Article En | MEDLINE | ID: mdl-21490378

INTRODUCTION: Recent genome wide association studies (GWAS) on coeliac disease (CD) have identified risk loci harbouring genes that fit the accepted pathogenic model and are considered aetiological candidates. METHODS: Using Taqman single nucleotide polymorphism (SNP) and expression assays, the study genotyped 11 SNPs tagging eight GWAS regions (1q31, 2q11-2q12, 3p21, 3q25-3q26, 3q28, 4q27, 6q25 and 12q24) in a Spanish cohort of 1094 CD patients and 540 controls, and performed expression analyses of candidate genes (RGS1, IL18R1/IL18RAP, CCR3, IL12A/SCHIP1, LPP, IL2/IL21-KIAA1109, TAGAP, and SH2B3) in intestinal mucosa from 29 CD children and eight controls. RESULTS: Polymorphisms in 1q31, 2q11-2q12, and 3q25 showed association in our cohort, and also 3q28 and 4q27 when combined with a previous study. Expression levels of IL12A, IL18RAP, IL21, KIAA1109, LPP, SCHIP1, and SH2B3 were affected by disease status, but the correlation between genotype and mRNA levels was observed only in IL12A, LPP, SCHIP1, and SH2B3. CONCLUSIONS: Expression differences between treated CD patients and controls along with SNP expression associations suggest a possible primary role for these four genes and their variants in pathogenesis. The lack of SNP effect in the remaining genes is probably a consequence of arbitrary candidate gene selection within association signals that are not based on functional studies.


Celiac Disease/genetics , Gene Expression Regulation , Genome-Wide Association Study , Alleles , Case-Control Studies , Female , Gene Expression Profiling , Genetic Loci , Genetic Predisposition to Disease , Humans , Male , Polymorphism, Single Nucleotide/genetics , Spain
...