Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.956
Filtrar
1.
Rev Assoc Med Bras (1992) ; 70(4): e20231490, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38716950

RESUMEN

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.


Asunto(s)
Autoinmunidad , Enfermedad Celíaca , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Polimorfismo de Nucleótido Simple , ARN Largo no Codificante , Humanos , Enfermedad Celíaca/genética , ARN Largo no Codificante/genética , Estudios de Casos y Controles , Niño , Polimorfismo de Nucleótido Simple/genética , Femenino , Masculino , Predisposición Genética a la Enfermedad/genética , Autoinmunidad/genética , Preescolar , Adolescente
2.
Sci Rep ; 14(1): 11528, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773317

RESUMEN

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.


Asunto(s)
Enfermedades Autoinmunes , Estudio de Asociación del Genoma Completo , Cirrosis Hepática Biliar , Análisis de la Aleatorización Mendeliana , Humanos , Cirrosis Hepática Biliar/genética , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/complicaciones , Colitis Ulcerosa/genética , Colitis Ulcerosa/complicaciones , Artritis Reumatoide/genética , Artritis Reumatoide/complicaciones , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/complicaciones , Síndrome de Sjögren/genética , Síndrome de Sjögren/complicaciones , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/complicaciones , Predisposición Genética a la Enfermedad , Enfermedad Celíaca/genética , Enfermedad Celíaca/complicaciones , Enfermedad de Graves/genética , Factores de Riesgo , Enfermedad de Crohn/genética , Enfermedad de Crohn/complicaciones , Esclerodermia Sistémica/genética , Esclerosis Múltiple/genética , Polimorfismo de Nucleótido Simple , Psoriasis/genética , Psoriasis/complicaciones
3.
PLoS One ; 19(5): e0301998, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38701071

RESUMEN

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.


Asunto(s)
COVID-19 , Enfermedad Celíaca , Microbioma Gastrointestinal , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , SARS-CoV-2 , Enfermedad Celíaca/genética , Enfermedad Celíaca/epidemiología , COVID-19/epidemiología , COVID-19/genética , COVID-19/virología , Humanos , Microbioma Gastrointestinal/genética , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/genética
4.
Medicine (Baltimore) ; 103(15): e37736, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38608121

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Sarcoidosis , Humanos , Enfermedad Celíaca/complicaciones , Enfermedad Celíaca/epidemiología , Enfermedad Celíaca/genética , Análisis de la Aleatorización Mendeliana , Sarcoidosis/epidemiología , Sarcoidosis/genética , Causalidad , Oportunidad Relativa
5.
Diabetes Res Clin Pract ; 211: 111683, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38663549

RESUMEN

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.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Diabetes Mellitus Tipo 1 , Obesidad , Humanos , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Diabetes Mellitus Tipo 1/genética , Femenino , Masculino , Adulto , Obesidad/genética , Proteínas/genética , Dislipidemias/genética , Dislipidemias/epidemiología , Comorbilidad , Persona de Mediana Edad , Predisposición Genética a la Enfermedad , Genotipo , Enfermedad Celíaca/genética , Enfermedad Celíaca/epidemiología , Hipertensión/genética , Hipertensión/epidemiología , Retinopatía Diabética/genética , Retinopatía Diabética/epidemiología , Polimorfismo de Nucleótido Simple , Adulto Joven
6.
Int Immunopharmacol ; 133: 112059, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38615385

RESUMEN

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.


Asunto(s)
Enfermedad Celíaca , Colitis Ulcerosa , Análisis de la Célula Individual , Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Colitis Ulcerosa/genética , Colitis Ulcerosa/inmunología , Humanos , Transcriptoma , Perfilación de la Expresión Génica , Análisis de Secuencia de ARN
8.
Clin Immunol ; 263: 110202, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38575045

RESUMEN

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.


Asunto(s)
Tirosina Quinasa del Receptor Axl , Enfermedad Celíaca , Duodeno , Péptidos y Proteínas de Señalización Intercelular , Mucosa Intestinal , Proteína S , Proteínas Tirosina Quinasas Receptoras , Tirosina Quinasa c-Mer , Humanos , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/inmunología , Masculino , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Femenino , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Adulto , Duodeno/metabolismo , Duodeno/inmunología , Duodeno/patología , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa c-Mer/metabolismo , Proteína S/metabolismo , Proteína S/genética , Persona de Mediana Edad , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Adulto Joven , Transducción de Señal , Adolescente , Interferones/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
9.
Iran Biomed J ; 28(1): 8-14, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-38444380

RESUMEN

Celiac disease (CD) is a complex disorder influenced by genetic and environmental factors. When people with a genetic predisposition to CD consume gluten, an inflammatory response is triggered in the small intestine, and this reaction can be alleviated by the elimination of gluten from the diet. The clinical manifestations of CD vary greatly from person to person and begin at a young age or in adulthood. Influence of genetic factors on CD development is evident in carriers of the DQ2 and/or DQ8 allele. HLA genotypes are associated with gut colonization by bacteria, particularly in individuals suffering from CD. In addition, beneficial gut microbes are crucial for the production of DPP-4, which plays a key role in immune function, as well as metabolic and intestinal health. Therefore, probiotics have been recommended as a complementary food supplement in CD.


Asunto(s)
Enfermedad Celíaca , Humanos , Enfermedad Celíaca/genética , Enfermedad Celíaca/terapia , Glútenes , Alelos , Predisposición Genética a la Enfermedad , Genotipo
10.
Acta Diabetol ; 61(6): 791-805, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38483572

RESUMEN

AIM: This study was undertaken to explicate the shared and distinctive genetic susceptibility and immune dysfunction in patients with T1D alone and T1D with CD (T1D + CD). METHODS: A total of 100 T1D, 50 T1D + CD and 150 healthy controls were recruited. HLA-DRB1/DQB1 alleles were determined by PCR-sequence-specific primer method, SNP genotyping for CTLA-4 and PTPN22 was done by simple probe-based SNP-array and genotyping for INS-23 Hph1 A/T was done by RFLP. Autoantibodies and cytokine estimation was done by ELISA. Immune-regulation was analysed by flow-cytometry. Clustering of autoantigen epitopes was done by epitope cluster analytical tool. RESULTS: Both T1D alone and T1D + CD had a shared association of DRB1*03:01, DRB1*04, DRB3*01:07/15 and DQB1*02. DRB3*01:07/15 confers the highest risk for T1D with relative risk of 11.32 (5.74-22.31). Non-HLA gene polymorphisms PTPN22 and INS could discriminate between T1D and T1D + CD. T1D + CD have significantly higher titers of autoantibodies, expression of costimulatory molecules on CD4 and CD8 cells, and cytokine IL-17A and TGF-ß1 levels compared to T1D patients. Epitopes from immunodominant regions of autoantigens of T1D and CD clustered together with 40% homology. CONCLUSION: Same HLA genes provide susceptibility for both T1D and CD. Non-HLA genes CTLA4, PTPN22 and INS provide further susceptibility while different polymorphisms in PTPN22 and INS can discriminate between T1D and T1D + CD. Epitope homology between autoantigens of two diseases further encourages the two diseases to occur together. The T1D + CD being more common in females along with co-existence of thyroid autoimmunity, and have more immune dysregulated state than T1D alone.


Asunto(s)
Autoantígenos , Enfermedad Celíaca , Diabetes Mellitus Tipo 1 , Predisposición Genética a la Enfermedad , Proteína Tirosina Fosfatasa no Receptora Tipo 22 , Humanos , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , India/epidemiología , Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Femenino , Masculino , Autoantígenos/inmunología , Autoantígenos/genética , Niño , Adolescente , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Adulto , Cadenas beta de HLA-DQ/genética , Autoanticuerpos/inmunología , Autoanticuerpos/sangre , Cadenas HLA-DRB1/genética , Adulto Joven , Polimorfismo de Nucleótido Simple , Preescolar , Antígeno CTLA-4/genética , Genotipo , Estudios de Casos y Controles
11.
Clinics (Sao Paulo) ; 79: 100317, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38432123

RESUMEN

OBJECTIVE: To evaluate the relationship between genetic haplotypes associated with celiac disease (Human Leucocyte Antigen [HLA] DQ2 and DQ8) with the diagnosis, clinical presentation, and location of endometriosis in Brazilian women. METHOD: A retrospective cross-sectional study, was conducted in a Tertiary hospital. PATIENTS: Women aged 18-50 years who underwent HLA-DQ2 and HLA-DQ8 haplotype analysis. INTERVENTION: The patients were divided into endometriosis and control groups and evaluated for symptoms; endometriosis location, American Society for Reproductive Medicine (ASRM) stage, and the presence of anti-tissue transglutaminase IgA (anti-TgA), HLA-DQ2, and HLA-DQ8 markers. RESULTS: A total of 434 consecutive patients with (n = 315) and without (n = 119) endometriosis were included. Pain and infertility were more frequent in the endometriosis group than in the control group. The presence of HLA-DQ2, HLA-DQ8, and anti-TgA was similar between both groups. The presence of HLA-DQ2 and HLA-DQ8 markers did not differ based on age, pain symptoms, ASRM stage, or endometriosis location. CONCLUSION: Although there are similarities in inflammatory markers and pathophysiology between celiac disease and endometriosis, this study found no significant associations in the presence of HLA-DQ2 or HLA-DQ8 haplotypes and endometriosis.


Asunto(s)
Enfermedad Celíaca , Endometriosis , Antígenos HLA-DQ , Humanos , Femenino , Endometriosis/genética , Estudios de Casos y Controles , Estudios Retrospectivos , Haplotipos , Enfermedad Celíaca/genética , Estudios Transversales , Dolor
12.
J Pediatr Gastroenterol Nutr ; 78(5): 1143-1148, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38477348

RESUMEN

OBJECTIVES: Autoantibodies against tissue transglutaminase (tTG) are serological markers of celiac disease. The aim was to study the applicability of human leukocyte antigen (HLA)-genotyping and tTG autoantibodies in the screening of celiac disease in a longitudinal birth cohort followed to age 15 years. METHODS: Included were 13,860 HLA-DQ-genotyped children at birth and previously invited to a screening at age 3 and 9 years, respectively. HLA-DQB1*02 and/or DQB1*03:02 (HLA-risk) children were compared with non-HLA-DQB1*02 and non-DQB1*03:02 (HLA-nonrisk) children. The present study reinvited 12,948/13,860 (93.4%) children at age 15 years of whom 1056/2374 (44.5%) participated in screening at both age 3 and 9 years. Both immunoglobulin A (IgA) and G (IgG) autoantibodies against tTG were analyzed separately in radiobinding assays. Persistently tTG autoantibody-positive children were examined with intestinal biopsy to confirm the diagnosis of celiac disease. RESULTS: At age 3 years, celiac disease was diagnosed in 56/1635 (3.4%) HLA-risk children compared with 0/1824 HLA-nonrisk children (p < 0.001). By age 9 years, celiac disease was diagnosed in 72/1910 (3.8%) HLA-risk children compared with 0/2167 HLA-nonrisk children (p < 0.001). Screening at age 15 years detected 14/1071 (1.3%) HLA-risk children positive for IgA-tTG and/or IgG-tTG of whom 12/1071 (1.1%) remained persistently positive. Among those, 10/1071 (0.9%, 95% confidence interval: 0.4%-1.7%) HLA-risk children were diagnosed with celiac disease compared with 0/1303 HLA-nonrisk children (p < 0.001) and 5/491 (1.0%) were negative in screenings at both 3 and 9 years of age. CONCLUSIONS: Screening for celiac disease needs to be performed at multiple timepoints to detect all cases but can be restricted to children at HLA-risk.


Asunto(s)
Autoanticuerpos , Enfermedad Celíaca , Proteínas de Unión al GTP , Inmunoglobulina A , Transglutaminasas , Humanos , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/genética , Niño , Preescolar , Transglutaminasas/inmunología , Estudios Longitudinales , Autoanticuerpos/sangre , Adolescente , Femenino , Masculino , Inmunoglobulina A/sangre , Proteínas de Unión al GTP/inmunología , Inmunoglobulina G/sangre , Proteína Glutamina Gamma Glutamiltransferasa 2 , Antígenos HLA-DQ/genética , Tamizaje Masivo/métodos , Genotipo , Cadenas beta de HLA-DQ/genética , Factores de Riesgo , Predisposición Genética a la Enfermedad
13.
Asian Pac J Cancer Prev ; 25(2): 647-652, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38415552

RESUMEN

OBJECTIVE: Celiac disease (CD) and colorectal cancer (CRC) are distinct gastrointestinal conditions with a debated association. This study aimed to evaluate the mRNA expression of CD4 and Foxp3 in tissue specimens of CD and CRC patients. The findings can provide valuable insights into the complex connection between these different gastrointestinal conditions. METHODS: Tissue samples from 100 CRC patients, 50 CD patients, and 50 healthy controls (HCs) were collected. RNA extraction, cDNA synthesis, and quantitative real-time PCR were performed. Statistical analysis was conducted using ANOVA and Pearson's correlation test. RESULT: CD4 mRNA expression was significantly higher in CRC patients compared to CD patients and HCs (P<0.0001 for both). Foxp3 mRNA expression was significantly higher in CD patients compared to CRC patients and HCs (P<0.0001 for both). Clinicopathological characteristics did not correlate significantly with gene expression levels. CONCLUSION: This study reveals differential expression patterns of CD4 and Foxp3 mRNA in CRC and CD patients. Upregulated CD4 mRNA suggests its potential role in promoting tumor growth, while increased Foxp3 mRNA expression may reflect an immunosuppressive mechanism in CD pathogenesis. These findings provide insights into the molecular and immunological aspects of CRC and CD, warranting further studies for potential therapeutic strategies.


Asunto(s)
Enfermedad Celíaca , Neoplasias Colorrectales , Humanos , Enfermedad Celíaca/genética , Enfermedad Celíaca/complicaciones , Enfermedad Celíaca/patología , Neoplasias Colorrectales/patología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Estudios de Casos y Controles , Proyectos de Investigación , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Immun Inflamm Dis ; 12(2): e1186, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38353316

RESUMEN

BACKGROUND: Celiac disease (CD) is a chronic autoimmune disorder characterized by an abnormal immune response to gluten, a protein found in wheat, barley, and rye. It is well established that the integrity of epithelial tight junctions (TJs) and adherens junctions (AJs) plays a crucial role in the pathogenesis of CD. These junctional complexes contribute to the apical-basal polarity of the intestinal epithelial cells, which is crucial for their proper functioning. METHODS: Sixty CD subjects, and 50 controls were enrolled in the current study. Mucosal samples were obtained from the distal duodenum, total RNA was extracted and complementary DNA was synthesized. The relative expression levels of the desired genes were evaluated by quantitative real-time polymerase chain reaction based on ΔΔCt method. The gene-gene interaction network was also constructed using GeneMANIA. RESULTS: CRB3 (p = .0005), LKB1 (p < .0001), and SCRIB (p = .0005) had lower expression in CD patients compared to controls, while PRKCZ expression did not differ between groups (p > .05). CRB3 represented a significant diagnostic value for differentiating CD patients from the control group (p = .02). CONCLUSION: The aim of the current study was to evaluate the changes in the mRNA expression levels of SCRIB, PRKCZ, LKB1, and CRB3 genes in the small intestinal biopsy samples of CD patients in comparison to the healthy control subjects. Our data uncover the importance of polarity-related genes (especially CRB3) in CD pahtomechanism, that may facilitate the planning of the future studies looking for finding innovative diagnostic and therapeutic strategies for CD.


Asunto(s)
Enfermedad Celíaca , Humanos , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/genética , Glútenes/metabolismo , Duodeno/metabolismo , Duodeno/patología , Biopsia , ARN Mensajero/genética , ARN Mensajero/metabolismo
15.
BMJ Open Gastroenterol ; 11(1)2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-38177066

RESUMEN

OBJECTIVE: The environmental trigger behind the increasing prevalence of coeliac disease is not known. One suggested cause is iron deficiency, which is common in coeliac disease. We aimed to evaluate this possible association with Mendelian randomisation (MR), which under certain assumptions can suggest a causal relationship. DESIGN: We conducted a two-sample MR study examining the relationship between single nucleotide polymorphisms (SNPs) associated with iron status and the presence of coeliac disease. The SNPs were drawn from a meta-analysis of three genome-wide association studies (GWAS). The association between these SNPs and coeliac disease was assessed using GWAS summary statistics from the UK Biobank. This consists of 336 638 white British individuals, 1855 with coeliac disease. We performed an MR Egger test for pleiotropy and assessed the plausibility of the assumptions of MR to evaluate for possible causality. RESULTS: There were four SNPs strongly associated with systemic iron status. These were not associated with known risk factors for coeliac disease. All four SNPs were available in the UK Biobank coeliac disease summary statistics. Harmonising exposure and outcome associations, we found that higher iron status was negatively associated with risk of coeliac disease (OR per 1 SD increase in serum iron: 0.65, 95% CI 0.47 to 0.91). Leave-one-out analyses had consistent results, and no single SNP drove the association. All three assumptions of MR appeared plausible. CONCLUSION: We found that genetically lower iron levels were associated with an increased risk of coeliac disease. Our findings highlight a potential opportunity for coeliac disease prevention.


Asunto(s)
Enfermedad Celíaca , Estudio de Asociación del Genoma Completo , Humanos , Enfermedad Celíaca/epidemiología , Enfermedad Celíaca/genética , Estudio de Asociación del Genoma Completo/métodos , Hierro , Polimorfismo de Nucleótido Simple , Factores de Riesgo
16.
Int J Immunogenet ; 51(2): 89-92, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38296826

RESUMEN

Celiac disease (CD) is an immune disorder, that is triggered by gluten ingestion in genetically predisposed individuals. The HLA-DQB1*02 allele is the main predisposing genetic factor and a candidate for first-line genotyping screening. We designed and validated a simple, DNA purification-free PCR protocol directly from crude saliva, enabling the detection of the DQB1*02 allele. This assay also distinguishes homozygous from heterozygous carriers. We propose this method for use in mass screening and/or epidemiological studies.


Asunto(s)
Enfermedad Celíaca , Saliva , Humanos , Genotipo , Cadenas beta de HLA-DQ/genética , Predisposición Genética a la Enfermedad , Alelos , Enfermedad Celíaca/genética , Enfermedad Celíaca/diagnóstico , Reacción en Cadena de la Polimerasa
17.
Int J Mol Sci ; 25(2)2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38255930

RESUMEN

We comprehensively evaluated the expression of therapeutically targetable immune checkpoint molecules involved in celiac disease (CD). We have focused on the alteration of the CD200/CD200R pathway and Elafin expression in celiac disease and discussed their roles in regulating the immune response. There are limited data related to the expression or function of these molecules in celiac disease. This finding could significantly contribute to the understanding of the clinical manifestation of CD. CD200, CD200R and Elafin distributions were determined by ELISA and immunohistochemistry analyses in serum and biopsies of CD patients. Analyses of Th1 and Th17 cytokines were determined. PCR amplification of a fragment of the PI3 gene was carried out using genomic DNA isolated from whole blood samples of the study subjects. Different aliquots of the PCR reaction product were subjected to RFLP analysis for SNP genotyping and detection. We characterized the expression and function of the CD200-CD200R axis and PI3 in celiac disease. A significantly higher level of soluble CD200 and CD200R and lower expression of PI3 in serum of CD patients was observed compared to healthy controls. Consistent with our results, CD200 expression is regulated by IFN-gamma. Interaction of CD200/CD200R leads to production of type-Th1 and -Th17 cytokines. Regarding the PI3 genotype, the CT genotype proportion SNP rs1733103 and the GG genotype SNP rs41282752 were predominant in CD patients. SNP rs1733103 showed a significant association between the SNP variables and CD. In celiac disease the immune checkpoint is compromised or dysregulated, which can contribute to inflammation and the autoimmunity process. The study of these checkpoint points will lead to the development of targeted therapies aimed at restoring immunological balance in CD. Specific coding regions of the PI3 gene-splice variants predispose the Elafin protein, both at the transcriptional and post-translational levels, to modify its expression and function, resulting in reduced differential functional protein levels in patients with active celiac disease.


Asunto(s)
Enfermedad Celíaca , Proteínas de Punto de Control Inmunitario , Humanos , Elafina , Enfermedad Celíaca/genética , Genotipo , Citocinas/genética
18.
Biochimie ; 219: 51-54, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37524198

RESUMEN

Clock gene disruption has been reported in inflammatory and autoimmune diseases. Specifically, it has been shown that clock gene expression is down-regulated in intestinal tissue and peripheral blood mononuclear cells of patients with inflammatory bowel disease (IBD). We aimed to determine the systemic expression of the circadian clock genes in newly diagnosed untreated, young patients with celiac disease (CeD). We prospectively enrolled patients younger than 20 years old who underwent diagnostic endoscopic procedures either for CeD diagnosis or due to other gastrointestinal complaints, at the pediatric and adult gastroenterology units, the Tel Aviv Sourasky Medical Center from 8/2016-8/2022. Demographic data, anthropometric parameters, and endoscopic macroscopic and microscopic findings were obtained. Blood samples were obtained to determine tissue transglutaminase (tTG) and core clock gene (CLOCK, BMAL1, PER1, PER2, CRY1, CRY2) expression in white blood cells (WBC). Thirty individuals were analyzed (18 with newly diagnosed CeD and 12 controls). Expression of the clock genes CLOCK, BMAL1, CRY2, PER1 and PER2 was significantly reduced in CeD patients compared to controls, while CRY1 did not differ between the groups. In conclusion, newly diagnosed, untreated, young patients with CeD have reduced clock gene expression in WBC compared to controls. These results suggest that, in CeD, the inflammatory response is associated with systemic disruption of clock gene expression, as is manifested in other inflammatory and autoimmune diseases. CLINICALTRIALS.GOV IDENTIFIER: NCT03662646.


Asunto(s)
Enfermedades Autoinmunes , Enfermedad Celíaca , Relojes Circadianos , Adulto , Humanos , Niño , Adulto Joven , Relojes Circadianos/genética , Ritmo Circadiano/genética , Leucocitos Mononucleares , Factores de Transcripción ARNTL/genética , Enfermedad Celíaca/genética
19.
Lancet Child Adolesc Health ; 8(1): 63-74, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37972632

RESUMEN

Paediatric prospective studies of coeliac disease with longitudinal collection of biological samples and clinical data offer a unique perspective on disease risk. This Review highlights the information now available from international paediatric prospective studies on genetic and environmental risk factors for coeliac disease. In addition, recent omics studies have made it possible to study complex interactions between genetic and environmental factors and thereby further our insight into the causes of the disease. In the future, paediatric prospective studies will be able to provide more detailed risk prediction models combining genes, the environment, and biological corroboration from multiomics. Such studies could also contribute to biomarker development and an improved understanding of disease pathogenesis.


Asunto(s)
Enfermedad Celíaca , Niño , Humanos , Enfermedad Celíaca/epidemiología , Enfermedad Celíaca/genética , Estudios Prospectivos , Biomarcadores
20.
Int J Immunogenet ; 51 Suppl 1: 3-20, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38153308

RESUMEN

Coeliac disease is a common immune-mediated inflammatory disorder caused by dietary gluten in genetically susceptible individuals. While the diagnosis of coeliac disease is based on serological and histological criteria, HLA-DQ genotyping can be useful, especially in excluding the diagnosis in patients who do not carry the relevant DQ heterodimers: DQA1*05 DQB1*02, DQB1*03:02 or DQA1*02 DQB1*02 (commonly referred to as DQ2.5, DQ8 and DQ2.2, respectively). External quality assessment results for HLA genotyping in coeliac disease have revealed concerning errors in HLA genotyping, reporting and clinical interpretation. In response, these guidelines have been developed as an evidence-based approach to guide laboratories undertaking HLA genotyping for coeliac disease and provide recommendations for reports to standardise and improve the communication of results.


Asunto(s)
Enfermedad Celíaca , Antígenos HLA-DQ , Humanos , Genotipo , Antígenos HLA-DQ/genética , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/genética , Predisposición Genética a la Enfermedad , Reino Unido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...