Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.969
Filter
1.
Adv Exp Med Biol ; 1449: 95-111, 2024.
Article in English | MEDLINE | ID: mdl-39060733

ABSTRACT

The incidence of chronic inflammatory diseases (CIDs) is dramatically increasing in the developed world, resulting in an increased burden of disease in childhood. Currently, there are limited effective strategies for treating or preventing these conditions. To date, myriads of cross-sectional studies have described alterations in the composition of the gut microbiota in a variety of disease states, after the disease has already occurred. We suggest that to mechanically link these microbiome changes with disease pathogenesis, a prospective cohort design is needed to capture changes that precede or coincide with disease onset and symptoms. In addition, these prospective studies must integrate microbiological, metagenomic, meta transcriptomic and metabolomic data with minimal and standardized clinical and environmental metadata that allow to correctly compare and interpret the results of the analysis of the human microbiota in order to build a system-level model of the interactions between the host and the development of the disease. The creation of new biological computational models thus constructed will allow us to finally move from the detection of simple elements of "association" to the identification of elements of real "causality" allowing to provide a mechanistic approach to the exploration of the development of CIDs.This can only be done when these diseases are studied as complex biological networks. In this chapter we discuss the current knowledge regarding the contribution of the microbiome to CID in childhood, focusing on celiac disease and inflammatory bowel disease, with the overall aim of identifying pathways to shift research from descriptive to mechanistic approaches. We then examine how some components of the microbiota, through epigenetic reprogramming, can start the march from genetic predisposition to clinical expression of CIDs, thus opening up new possibilities for intervention, through microbiota therapy targeting the manipulation of the composition and function of the microbiota, for future applications of precision medicine and primary prevention.


Subject(s)
Celiac Disease , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Probiotics , Humans , Celiac Disease/microbiology , Celiac Disease/genetics , Celiac Disease/prevention & control , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/prevention & control , Probiotics/therapeutic use
2.
Genes (Basel) ; 15(7)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39062631

ABSTRACT

Celiac disease (CD) is a complicated autoimmune disease that is caused by gluten sensitivity. It was commonly believed that CD only affected white Europeans, but recent findings show that it is also prevailing in some other racial groups, like South Asians, Caucasians, Africans, and Arabs. Genetics plays a profound role in increasing the risk of developing CD. Genetic Variations in non-HLA genes such as LPP, ZMIZ1, CCR3, and many more influence the risk of CD in various populations. This study aimed to explore the association between LPP rs1464510 and ZMIZ1 rs1250552 and CD in the Punjabi Pakistani population. For this, a total of 70 human subjects were selected and divided into healthy controls and patients. Genotyping was performed using an in-house-developed tetra-amplification refractory mutation system polymerase chain reaction. Statistical analysis revealed a significant association between LPP rs1464510 (χ2 = 4.421, p = 0.035) and ZMIZ1 rs1250552 (χ2 = 3.867, p = 0.049) and CD. Multinomial regression analysis showed that LPP rs1464510 A allele reduces the risk of CD by ~52% (OR 0.48, CI: 0.24-0.96, 0.037), while C allele-carrying subjects are at ~2.6 fold increased risk of CD (OR 3.65, CI: 1.25-10.63, 0.017). Similarly, the ZMIZ1 rs1250552 AG genotype significantly reduces the risk of CD by 73% (OR 0.26, CI: 0.077-0.867, p = 0.028). In summary, Genetic Variations in the LPP and ZMIZ1 genes influence the risk of CD in Punjabi Pakistani subjects. LPP rs1464510 A allele and ZMIZ1 AG genotype play a protective role and reduce the risk of CD.


Subject(s)
Celiac Disease , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Humans , Celiac Disease/genetics , Pakistan , Male , Female , Transcription Factors/genetics , Adult , Case-Control Studies , Alleles , Genotype , Child , Adolescent
4.
Ren Fail ; 46(2): 2357246, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38832490

ABSTRACT

OBJECTIVE: A two-sample Mendelian randomization (MR) analysis was performed to elucidate the causal impact of celiac disease on the risk of chronic kidney disease (CKD). METHODS: The study comprised data from three genome-wide association studies involving individuals of European ancestry. The study groups included participants with celiac disease (n = 24,269), CKD (n = 117,165), and estimated glomerular filtration rate levels based on serum creatinine (eGFRcrea, n = 133,413). We employed four widely recognized causal inference algorithms: MR-Egger, inverse variance weighted (IVW), weighted median, and weighted mode. To address potential issues related to pleiotropy and overall effects, MR-Egger regression and the MR-PRESSO global test were performed. Heterogeneity was assessed using Cochran's Q test. RESULTS: We identified 14 genetic variants with genome-wide significance. The MR analysis provided consistent evidence across the various methodologies, supporting a causal relationship between celiac disease and an elevated risk of CKD (odds ratio (OR)IVW = 1.027, p = 0.025; ORweighted median = 1.028, P = 0.049; ORweighted mode = 1.030, p = 0.044). Furthermore, we observed a causal link between celiac disease and a decreased eGFRcrea (ORIVW = 0.997, P = 2.94E-06; ORweighted median = 0.996, P = 1.68E-05; ORweighted mode = 0.996, P = 3.11E-04; ORMR Egger = 0.996, P = 5.00E-03). We found no significant evidence of horizontal pleiotropy, heterogeneity, or bias based on MR-Egger regression, MR-PRESSO, and Cochran's Q test. CONCLUSION: The results of this study indicate a causal relationship between celiac disease and an increased risk of CKD.


Subject(s)
Celiac Disease , Genome-Wide Association Study , Glomerular Filtration Rate , Mendelian Randomization Analysis , Renal Insufficiency, Chronic , Humans , Celiac Disease/genetics , Celiac Disease/complications , Renal Insufficiency, Chronic/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Female , Male , Risk Factors
5.
Medicine (Baltimore) ; 103(25): e38474, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905357

ABSTRACT

The link between celiac disease (CeD) and thyroid dysfunction has been investigated. However, it is uncertain if CeD is causally linked to thyroid dysfunction. A 2-sample Mendelian randomization study was conducted to ascertain the causal connection between CeD and thyroid dysfunction. Using data from the FinnGen Consortium, a 2-sample Mendelian randomization study was conducted to look at the connection between thyroid dysfunction and CeD. Another replication of the data from the UK Biobank was subsequently performed to confirm our findings. Furthermore, a sequence of sensitivity analyses was performed. The inverse variance weighting technique demonstrates that genetically determined CeD is substantially linked with hypothyroidism, thyrotoxicosis, Graves' disease, and free thyroxine. However, no significant associations were found between CeD and thyroid-stimulating hormone or thyroiditis. Moreover, we achieve the same results in duplicate datasets, which increases the reliability of our findings. This study suggests that CeD and thyroid dysfunction are linked, and it gives theoretical support and new ways of thinking about how to diagnose and treat both conditions.


Subject(s)
Celiac Disease , Mendelian Randomization Analysis , Thyroid Diseases , Humans , Celiac Disease/genetics , Celiac Disease/complications , Celiac Disease/epidemiology , Thyroid Diseases/genetics , Thyroid Diseases/epidemiology , Hypothyroidism/genetics , Hypothyroidism/epidemiology , Thyrotropin/blood
6.
Gene ; 927: 148681, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38871036

ABSTRACT

RNA sequencing (RNA-seq) has undergone substantial advancements in recent decades and has emerged as a vital technique for profiling the transcriptome. The transition from bulk sequencing to single-cell and spatial approaches has facilitated the achievement of higher precision at cell resolution. It provides valuable biological knowledge about individual immune cells and aids in the discovery of the molecular mechanisms that contribute to the development of autoimmune diseases. Celiac disease (CeD) is an autoimmune disorder characterized by a strong immune response to gluten consumption. RNA-seq has led to significantly advanced research in multiple fields, particularly in CeD research. It has been instrumental in studies involving comparative transcriptomics, nutritional genomics and wheat research, cancer research in the context of CeD, genetic and noncoding RNA-mediated epigenetic insights, disease monitoring and biomarker discovery, regulation of mitochondrial functions, therapeutic target identification and drug mechanism of action, dietary factors, immune cell profiling and the immune landscape. This review offers a comprehensive examination of recent RNA-seq technology research in the field of CeD, highlighting future challenges and opportunities for its application.


Subject(s)
Celiac Disease , Celiac Disease/genetics , Celiac Disease/immunology , Humans , Sequence Analysis, RNA/methods , Transcriptome , Gene Expression Profiling/methods , Epigenesis, Genetic
8.
Viruses ; 16(5)2024 04 26.
Article in English | MEDLINE | ID: mdl-38793567

ABSTRACT

Directed evolution is a pivotal strategy for new antibody discovery, which allowed the generation of high-affinity Fabs against gliadin from two antibody libraries in our previous studies. One of the libraries was exclusively derived from celiac patients' mRNA (immune library) while the other was obtained through a protein engineering approach (semi-immune library). Recent advances in high-throughput DNA sequencing techniques are revolutionizing research across genomics, epigenomics, and transcriptomics. In the present work, an Oxford Nanopore in-lab sequencing device was used to comprehensively characterize the composition of the constructed libraries, both at the beginning and throughout the phage-mediated selection processes against gliadin. A customized analysis pipeline was used to select high-quality reads, annotate chain distribution, perform sequence analysis, and conduct statistical comparisons between the different selection rounds. Some immunological attributes of the most representative phage variants after the selection process were also determined. Sequencing results revealed the successful transfer of the celiac immune response features to the immune library and the antibodies derived from it, suggesting the crucial role of these features in guiding the selection of high-affinity recombinant Fabs against gliadin. In summary, high-throughput DNA sequencing has improved our understanding of the selection processes aimed at generating molecular binders against gliadin.


Subject(s)
Gliadin , High-Throughput Nucleotide Sequencing , Immunoglobulin Fab Fragments , Nanopore Sequencing , Peptide Library , Gliadin/immunology , Gliadin/genetics , Humans , High-Throughput Nucleotide Sequencing/methods , Nanopore Sequencing/methods , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Celiac Disease/immunology , Celiac Disease/genetics , Cell Surface Display Techniques/methods
9.
J Clin Immunol ; 44(6): 133, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38780872

ABSTRACT

PURPOSE: A large proportion of Common variable immunodeficiency (CVID) patients has duodenal inflammation with increased intraepithelial lymphocytes (IEL) of unknown aetiology. The histologic similarities to celiac disease, lead to confusion regarding treatment (gluten-free diet) of these patients. We aimed to elucidate the role of epigenetic DNA methylation in the aetiology of duodenal inflammation in CVID and differentiate it from true celiac disease. METHODS: DNA was isolated from snap-frozen pieces of duodenal biopsies and analysed for differences in genome-wide epigenetic DNA methylation between CVID patients with increased IEL (CVID_IEL; n = 5) without IEL (CVID_N; n = 3), celiac disease (n = 3) and healthy controls (n = 3). RESULTS: The DNA methylation data of 5-methylcytosine in CpG sites separated CVID and celiac diseases from healthy controls. Differential methylation in promoters of genes were identified as potential novel mediators in CVID and celiac disease. There was limited overlap of methylation associated genes between CVID_IEL and Celiac disease. High frequency of differentially methylated CpG sites was detected in over 100 genes nearby transcription start site (TSS) in both CVID_IEL and celiac disease, compared to healthy controls. Differential methylation of genes involved in regulation of TNF/cytokine production were enriched in CVID_IEL, compared to healthy controls. CONCLUSION: This is the first study to reveal a role of epigenetic DNA methylation in the etiology of duodenal inflammation of CVID patients, distinguishing CVID_IEL from celiac disease. We identified potential biomarkers and therapeutic targets within gene promotors and in high-frequency differentially methylated CpG regions proximal to TSS in both CVID_IEL and celiac disease.


Subject(s)
Celiac Disease , Common Variable Immunodeficiency , CpG Islands , DNA Methylation , Duodenum , Epigenesis, Genetic , Humans , Common Variable Immunodeficiency/genetics , Duodenum/metabolism , Duodenum/pathology , Celiac Disease/genetics , Female , Male , Adult , Middle Aged , CpG Islands/genetics , Promoter Regions, Genetic/genetics , Intraepithelial Lymphocytes/immunology , Young Adult , Genome-Wide Association Study , 5-Methylcytosine/metabolism
10.
Clin Transl Gastroenterol ; 15(7): e00710, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38713138

ABSTRACT

INTRODUCTION: Celiac disease (CD) and type 1 diabetes (T1D) often co-occur and share genetic components in the human leukocyte antigen (HLA) class II region. We aimed to study the usefulness of HLA genotyping in predicting the risk of developing T1D in patients with CD and the temporal relationship between these diseases. METHODS: A cohort of 1,886 Sardinian patients, including 822 with CD, 1,064 with T1D, and 627 controls, underwent HLA class II typing. Seventy-six of 822 patients with CD were also affected by T1D (CD-T1D), and their HLA genotypes were analyzed for specific HLA associations with CD, T1D, and controls. RESULTS: High-risk HLA-DQ genotypes, including HLA-DQ2.5/DQ8, -DQ2.5/DQ2.5, and -DQ2.5/DQ2.3, were strongly associated with CD-T1D with frequencies of 34.5%, 15.9%, and 18.8%, respectively. Conversely, certain HLA genotypes associated with CD seemed to confer protection against T1D development. Therefore, HLA genotyping allows for the identification of those patients with CD who might develop T1D. The frequency of patients with CD preceding T1D is higher in younger children than older ones, with implications for the early childhood approach to diabetes prevention. DISCUSSION: CD is a condition for future T1D development, and specific HLA genotypes can predict this risk. Early screening for celiac autoimmunity and subsequent HLA typing in CD children could help identify those at high risk of T1D, allowing for proactive interventions and immunotherapies to preserve ß-cell function. These findings may support the re-evaluation of HLA typing in children with CD.


Subject(s)
Celiac Disease , Diabetes Mellitus, Type 1 , Genetic Predisposition to Disease , Genotype , HLA-DQ Antigens , Humans , Celiac Disease/genetics , Celiac Disease/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Child , Male , Female , Child, Preschool , HLA-DQ Antigens/genetics , Adolescent , Italy/epidemiology , Infant , Risk Factors , Case-Control Studies
11.
Sci Rep ; 14(1): 11528, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773317

ABSTRACT

As an autoimmune disease, up to 73% of patients with primary biliary cholangitis (PBC) have a combination of extrahepatic autoimmune diseases (EHAIDs); however, the causal relationship between PBC and EHAIDs is unclear. The genome-wide association analyses provided 14 GWAS data for PBC and EHAIDs, and bidirectional, two-sample MR analyses were performed to examine the relationship between PBC and EHAIDs. The analysis using MR provides a strong and meaningful estimation of the bidirectional correlation between PBC and 7 EHAIDs: rheumatoid arthritis, systemic lupus erythematosus, Sjögren's syndrome, systemic sclerosis, autoimmune hypothyroidism, inflammatory bowel disease and ulcerative colitis of its types. In addition, PBC increases the risk of autoimmune thyroid diseases such as autoimmune hyperthyroidism and Graves' disease, as well as multiple sclerosis and psoriasis. Additionally, PBC is identified as a risk factor for Crohn's disease and Celiac disease. Based on genetic evidence, there may be connections between PBC and specific EHAIDs: not all coexisting EHAIDs induce PBC, and vice versa. This underscores the significance of prioritizing PBC in clinical practice. Additionally, if any liver function abnormalities are observed during treatment or with EHAIDs, it is crucial to consider the possibility of comorbid PBC.


Subject(s)
Autoimmune Diseases , Genome-Wide Association Study , Liver Cirrhosis, Biliary , Mendelian Randomization Analysis , Humans , Liver Cirrhosis, Biliary/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/complications , Colitis, Ulcerative/genetics , Colitis, Ulcerative/complications , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/complications , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/complications , Sjogren's Syndrome/genetics , Sjogren's Syndrome/complications , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/complications , Genetic Predisposition to Disease , Celiac Disease/genetics , Celiac Disease/complications , Graves Disease/genetics , Risk Factors , Crohn Disease/genetics , Crohn Disease/complications , Scleroderma, Systemic/genetics , Multiple Sclerosis/genetics , Polymorphism, Single Nucleotide , Psoriasis/genetics , Psoriasis/complications
12.
Rev Assoc Med Bras (1992) ; 70(4): e20231490, 2024.
Article in English | MEDLINE | ID: mdl-38716950

ABSTRACT

OBJECTIVE: Various studies have reported that certain long non-coding RNA levels are unusually low in the intestines of celiac disease patients, suggesting that this may be associated with the inflammation observed in celiac disease. Despite these studies, the research aimed at uncovering the potential role of long non-coding RNAs in the pathogenesis of autoimmune diseases like celiac disease remains insufficient. Therefore, in this study, we plan to assess long non-coding RNA polymorphisms associated with autoimmunity in children diagnosed with celiac disease according to the European Society for Paediatric Gastroenterology Hepatology and Nutrition criteria. METHODS: DNA was isolated from paraffin tissue samples of 88 pediatric celiac disease patients and 74 healthy pediatric individuals. Single-nucleotide polymorphism genotyping of five long non-coding RNA polymorphisms associated with autoimmunity (LINC01934-rs1018326, IL18RAP-rs917997, AP002954.4-rs10892258, UQCRC2P1-rs6441961, and HCG14 rs3135316) was conducted using the TaqMan single-nucleotide polymorphism genotyping assays with the LightCycler 480. RESULTS: In our study, the genotypic and allelic frequency distribution of LINC01934-rs1018326 and AP002954.4-rs10892258 polymorphisms was found to be statistically significant in the comparison between the two groups (p<0.05). According to the multiple genetic model analyses, the LINC01934-rs1018326 polymorphism was observed to confer a 1.14-fold risk in the recessive model and a 1.2-fold risk in the additive model for pediatric celiac disease. Similarly, the AP002954.4-rs10892258 polymorphism was found to pose a 1.40-fold risk in the dominant model and a 1.7-fold risk in the additive model. CONCLUSION: Our study results draw attention to the LINC01934-rs1018326 and AP002954.4-rs10892258 polymorphisms in celiac disease and suggest that these polymorphisms may be associated with inflammation in autoimmune diseases like celiac disease.


Subject(s)
Autoimmunity , Celiac Disease , Gene Frequency , Genetic Predisposition to Disease , Genotype , Polymorphism, Single Nucleotide , RNA, Long Noncoding , Humans , Celiac Disease/genetics , RNA, Long Noncoding/genetics , Case-Control Studies , Child , Polymorphism, Single Nucleotide/genetics , Female , Male , Genetic Predisposition to Disease/genetics , Autoimmunity/genetics , Child, Preschool , Adolescent
13.
PLoS One ; 19(5): e0301998, 2024.
Article in English | MEDLINE | ID: mdl-38701071

ABSTRACT

Celiac disease exhibits a higher prevalence among patients with coronavirus disease 2019. However, the potential influence of COVID-19 on celiac disease remains uncertain. Considering the significant association between gut microbiota alterations, COVID-19 and celiac disease, the two-step Mendelian randomization method was employed to investigate the genetic causality between COVID-19 and celiac disease, with gut microbiota as the potential mediators. We employed the genome-wide association study to select genetic instrumental variables associated with the exposure. Subsequently, these variables were utilized to evaluate the impact of COVID-19 on the risk of celiac disease and its potential influence on gut microbiota. Employing a two-step Mendelian randomization approach enabled the examination of potential causal relationships, encompassing: 1) the effects of COVID-19 infection, hospitalized COVID-19 and critical COVID-19 on the risk of celiac disease; 2) the influence of gut microbiota on celiac disease; and 3) the mediating impact of the gut microbiota between COVID-19 and the risk of celiac disease. Our findings revealed a significant association between critical COVID-19 and an elevated risk of celiac disease (inverse variance weighted [IVW]: P = 0.035). Furthermore, we observed an inverse correlation between critical COVID-19 and the abundance of Victivallaceae (IVW: P = 0.045). Notably, an increased Victivallaceae abundance exhibits a protective effect against the risk of celiac disease (IVW: P = 0.016). In conclusion, our analysis provides genetic evidence supporting the causal connection between critical COVID-19 and lower Victivallaceae abundance, thereby increasing the risk of celiac disease.


Subject(s)
COVID-19 , Celiac Disease , Gastrointestinal Microbiome , Genome-Wide Association Study , Mendelian Randomization Analysis , SARS-CoV-2 , Celiac Disease/genetics , Celiac Disease/epidemiology , COVID-19/epidemiology , COVID-19/genetics , COVID-19/virology , Humans , Gastrointestinal Microbiome/genetics , SARS-CoV-2/isolation & purification , SARS-CoV-2/genetics
15.
Medicine (Baltimore) ; 103(15): e37736, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38608121

ABSTRACT

Observational research shows a link between celiac disease (CeD) and sarcoidosis, but the causal link between CeD and sarcoidosis is still unknown. A two-sample Mendelian randomization (MR) study was conducted to ascertain the causal connection between the 2 disorders. In our two-sample MR analysis, we identified independent genetic variants associated with CeD using publicly accessible GWAS data from people of European ancestry. Summary data for sarcoidosis were obtained from the FinnGen Consortium, the UK-Biobank, and a large GWAS dataset. To assess the association between CeD and sarcoidosis, our MR analysis used inverse variance weighted (IVW) as the primary method, incorporating the MR-Egger, weighted median (WM), and MR-PRESSO (outliers test) as a complementary method. In order to ensure that the findings were reliable, several sensitivity analyses were performed. Our study indicated that CeD had a significant causal relationship with sarcoidosis (IVW odds ratio (OR) = 1.13, 95% confidence interval (CI): 1.07-1.20, P = 5.58E-05; WM OR = 1.12, 95% CI: 1.03-1.23, P = 1.03E-02; MR-Egger OR = 1.07, 95% CI: 0.96-1.19, P = 2.20E-01). Additionally, we obtain the same results in the duplicated datasets as well, which makes our results even more reliable. The results of this investigation did not reveal any evidence of horizontal pleiotropy or heterogeneity. Our MR analysis showed a causal effect between CeD and an elevated risk of sarcoidosis. Further study is still needed to confirm the findings and look into the processes underlying these relationships.


Subject(s)
Celiac Disease , Sarcoidosis , Humans , Celiac Disease/complications , Celiac Disease/epidemiology , Celiac Disease/genetics , Mendelian Randomization Analysis , Sarcoidosis/epidemiology , Sarcoidosis/genetics , Causality , Odds Ratio
16.
Gastroenterology ; 167(1): 4-22, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670280

ABSTRACT

Celiac disease (CeD) is a gluten-induced enteropathy that develops in genetically susceptible individuals upon consumption of cereal gluten proteins. It is a unique and complex immune disorder to study as the driving antigen is known and the tissue targeted by the immune reaction can be interrogated. This review integrates findings gained from genetic, biochemical, and immunologic studies, which together have revealed mechanisms of gluten peptide modification and HLA binding, thereby enabling a maladapted anti-gluten immune response. Observations in human samples combined with experimental mouse models have revealed that the gluten-induced immune response involves CD4+ T cells, cytotoxic CD8+ T cells, and B cells; their cross-talks are critical for the tissue-damaging response. The emergence of high-throughput technologies is increasing our understanding of the phenotype, location, and presumably function of the gluten-specific cells, which are all required to identify novel therapeutic targets and strategies for CeD.


Subject(s)
Celiac Disease , Genetic Predisposition to Disease , Glutens , Celiac Disease/immunology , Celiac Disease/genetics , Humans , Glutens/immunology , Glutens/adverse effects , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology
17.
Clin Chem Lab Med ; 62(9): 1814-1823, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-38639193

ABSTRACT

OBJECTIVES: Non-celiac wheat sensitivity (NCWS) is an emerging clinical condition characterized by gastrointestinal and extraintestinal symptoms following the ingestion of gluten-containing foods in patients without celiac disease (CD) or wheat allergy. Despite the great interest for NCWS, the genetic risk factors still need to be fully clarified. In this study, we first assessed the possible contribution of KIR genes and KIR haplotypes on the genetic predisposition to NCWS. METHODS: Fifty patients with NCWS, 50 patients with CD, and 50 healthy controls (HC) were included in this study. KIR genes and KIR genotyping were investigated in all subjects by polymerase chain reaction with the sequence oligonucleotide probe (PCR-SSOP) method using Luminex technology. RESULTS: We found a statistically different distribution of some KIR genes among NCWS, CD, and HC. Specifically, NCWS showed a decreased frequency of KIR2DL1, -2DL3, -2DL5, -2DS2, -2DS3, -2DS4, -2DS5, and -3DS1 genes, and an increased frequency of -3DL1 gene respect to both CD and HC. No difference was detected in the KIR haplotype expression. At the multivariate analysis, KIR2DL5, -2DS4, and -2DS5 were independent predictors of NCWS. CONCLUSIONS: Our findings suggest a role of KIR genes in NCWS susceptibility, with KIR2DL5, -2DS4, and -2DS5 having a protective effect. Further large-scale multicentric studies are required to validate these preliminary findings.


Subject(s)
Genetic Predisposition to Disease , Haplotypes , Receptors, KIR , Wheat Hypersensitivity , Humans , Receptors, KIR/genetics , Female , Male , Adult , Wheat Hypersensitivity/genetics , Middle Aged , Celiac Disease/genetics , Case-Control Studies , Triticum/genetics , Genotype , Young Adult
18.
Diabetes Res Clin Pract ; 211: 111683, 2024 May.
Article in English | MEDLINE | ID: mdl-38663549

ABSTRACT

BACKGROUND AND AIMS: Because FTO gene is connected with the risk of obesity, cardiovascular disease and hypertension, as well as type 2 diabetes, we hypothesize that the rs9939609 FTO polymorphism may affect type 1 diabetes (T1D) complications and comorbidities. METHODS: We have investigated the associations of the FTO gene variant with the T1D and its complications and comorbidities, as well as the serum levels of pro- and anti-inflammatory markers and lipid profiles. RESULTS: The key results of our study are as follows: (1) the rs9939609 FTO polymorphism does not predispose individuals to T1D; (2) AA genotype is associated with an increased risk of overweight and obesity, retinopathy, hypertension, dyslipidemia and celiac disease; (3) AT genotype is associated with a decreased risk of retinopathy and celiac disease, whereas TT genotype is connected with decreased risk of dyslipidemia; (4) the FTO rs9939609 polymorphism affects the inflammatory status as well as lipid profile in T1D patients. CONCLUSIONS: Our results, for the first time, comprehensively indicate that the rs9939609 FTO polymorphism could be considered a genetic marker for increased susceptibility to T1D complications and comorbidities as well as suggests importance of FTO-mediated pathways in their etiology.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Diabetes Mellitus, Type 1 , Obesity , Humans , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Diabetes Mellitus, Type 1/genetics , Female , Male , Adult , Obesity/genetics , Proteins/genetics , Dyslipidemias/genetics , Dyslipidemias/epidemiology , Comorbidity , Middle Aged , Genetic Predisposition to Disease , Genotype , Celiac Disease/genetics , Celiac Disease/epidemiology , Hypertension/genetics , Hypertension/epidemiology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/epidemiology , Polymorphism, Single Nucleotide , Young Adult
19.
Int Immunopharmacol ; 133: 112059, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38615385

ABSTRACT

Many immune-mediated diseases have the common genetic basis, as an autoimmune disorder, celiac disease (CeD) primarily affects the small intestine, and is caused by the ingestion of gluten in genetically susceptible individuals. As for ulcerative colitis (UC), which most likely involves a complex interplay between some components of the commensal microbiota and other environmental factors in its origin. These two autoimmune diseases share a specific target organ, the bowel. The etiology and immunopathogenesis of both conditions characterized by chronic intestinal inflammation, ulcerative colitis and celiac disease, are not completely understood. Both are complex diseases with genetics and the environmental factors contributing to dysregulation of innate and adaptive immune responses, leading to chronic inflammation and disease. This study is designed to further clarify the relationship between UC and CeD. The GEO database was used to download gene expression profiles for CeD (GSE112102) and UC (GSE75214). The GSEA KEGG pathway analysis revealed that immune-related pathways were significantly associated with both diseases. Further, we screened 187 shared differentially expressed genes (DEGs) of the two diseases. Gene Ontology (GO) and WikiPathways were carried out to perform the biological process and pathway enrichment analysis. Subsequently, based on the DEGs, the least absolute shrinkage and selection operator (LASSO) analysis was performed to screen for the diagnostic biomarkers of the diseases. Moreover, single-cell RNA-sequencing (RNA-seq) data from five colonic propria with UC showed that REG4 expression was present in Goblet cell, Enteroendocrine cell, and Epithelial. Finally, our work identified REG4 is the shared gene of UC and CeD via external data validation, cellular experiments, and immunohistochemistry. In conclusion, our study elucidated that abnormal immune response could be the common pathogenesis of UC and CeD, and REG4 might be a key potential biomarker and therapeutic target for the comorbidity of these two diseases.


Subject(s)
Celiac Disease , Colitis, Ulcerative , Single-Cell Analysis , Celiac Disease/genetics , Celiac Disease/immunology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Humans , Transcriptome , Gene Expression Profiling , Sequence Analysis, RNA
20.
Clin Immunol ; 263: 110202, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38575045

ABSTRACT

Celiac disease (CD) is an immune-driven disease characterized by tissue damage in the small intestine of genetically-susceptible individuals. We evaluated here a crucial immune regulatory pathway involving TYRO3, AXL, and MERTK (TAM) receptors and their ligands PROS1 and GAS6 in duodenal biopsies of controls and CD patients. We found increased GAS6 expression associated with downregulation of PROS1 and variable TAM receptors levels in duodenum tissue of CD patients. Interestingly, CD3+ lymphocytes, CD68+, CD11c+ myeloid and epithelial cells, showed differential expressions of TAM components comparing CD vs controls. Principal component analysis revealed a clear segregation of two groups of CD patients based on TAM components and IFN signaling. In vitro validation demonstrated that monocytes, T lymphocytes and epithelial cells upregulated TAM components in response to IFN stimulation. Our findings highlight a dysregulated TAM axis in CD related to IFN signaling and contribute to a deeper understanding of the pathophysiology of CD.


Subject(s)
Axl Receptor Tyrosine Kinase , Celiac Disease , Duodenum , Intercellular Signaling Peptides and Proteins , Intestinal Mucosa , Protein S , Receptor Protein-Tyrosine Kinases , c-Mer Tyrosine Kinase , Humans , Celiac Disease/immunology , Celiac Disease/metabolism , Celiac Disease/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Male , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Female , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Adult , Duodenum/metabolism , Duodenum/immunology , Duodenum/pathology , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism , Protein S/metabolism , Protein S/genetics , Middle Aged , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Young Adult , Signal Transduction , Adolescent , Interferons/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL