Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nat Genet ; 56(5): 838-845, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38741015

RESUMEN

Autoimmune and inflammatory diseases are polygenic disorders of the immune system. Many genomic loci harbor risk alleles for several diseases, but the limited resolution of genetic mapping prevents determining whether the same allele is responsible, indicating a shared underlying mechanism. Here, using a collection of 129,058 cases and controls across 6 diseases, we show that ~40% of overlapping associations are due to the same allele. We improve fine-mapping resolution for shared alleles twofold by combining cases and controls across diseases, allowing us to identify more expression quantitative trait loci driven by the shared alleles. The patterns indicate widespread sharing of pathogenic mechanisms but not a single global autoimmune mechanism. Our approach can be applied to any set of traits and is particularly valuable as sample collections become depleted.


Asunto(s)
Alelos , Enfermedades Autoinmunes , Mapeo Cromosómico , Predisposición Genética a la Enfermedad , Sitios de Carácter Cuantitativo , Humanos , Enfermedades Autoinmunes/genética , Polimorfismo de Nucleótido Simple , Estudio de Asociación del Genoma Completo , Estudios de Casos y Controles , Herencia Multifactorial/genética
2.
Nat Med ; 29(6): 1520-1529, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37322120

RESUMEN

Primary sclerosing cholangitis (PSC) is an immune-mediated disease of the bile ducts that co-occurs with inflammatory bowel disease (IBD) in almost 90% of cases. Colorectal cancer is a major complication of patients with PSC and IBD, and these patients are at a much greater risk compared to patients with IBD without concomitant PSC. Combining flow cytometry, bulk and single-cell transcriptomics, and T and B cell receptor repertoire analysis of right colon tissue from 65 patients with PSC, 108 patients with IBD and 48 healthy individuals we identified a unique adaptive inflammatory transcriptional signature associated with greater risk and shorter time to dysplasia in patients with PSC. This inflammatory signature is characterized by antigen-driven interleukin-17A (IL-17A)+ forkhead box P3 (FOXP3)+ CD4 T cells that express a pathogenic IL-17 signature, as well as an expansion of IgG-secreting plasma cells. These results suggest that the mechanisms that drive the emergence of dysplasia in PSC and IBD are distinct and provide molecular insights that could guide prevention of colorectal cancer in individuals with PSC.


Asunto(s)
Colangitis Esclerosante , Neoplasias Colorrectales , Enfermedades Inflamatorias del Intestino , Humanos , Colangitis Esclerosante/complicaciones , Colangitis Esclerosante/patología , Inflamación/complicaciones , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/patología , Neoplasias Colorrectales/patología
3.
Trends Genet ; 39(4): 268-284, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36746737

RESUMEN

Genome-wide association studies (GWAS) have now correlated hundreds of genetic variants with complex genetic diseases and drug efficacy. Functional characterization of these factors remains challenging, particularly because of the lack of human model systems. Molecular and nanotechnological advances, in particular the ability to generate patient-specific PSC lines, differentiate them into diverse cell types, and seed and combine them on microfluidic chips, have led to the establishment of organ-on-a-chip (OoC) platforms that recapitulate organ biology. OoC technology thus provides unique personalized platforms for studying the effects of host genetics and environmental factors on organ physiology. In this review we describe the technology and provide examples of how OoCs may be used for disease modeling and pharmacogenetic research.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Sistemas Microfisiológicos , Farmacogenética , Estudio de Asociación del Genoma Completo , Genética Humana
4.
Front Immunol ; 13: 843086, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35371081

RESUMEN

Celiac Disease (CeD) is a complex immune disorder involving villous atrophy in the small intestine that is triggered by gluten intake. Current CeD diagnosis is based on late-stage pathophysiological parameters such as detection of specific antibodies in blood and histochemical detection of villus atrophy and lymphocyte infiltration in intestinal biopsies. To date, no early onset biomarkers are available that would help prevent widespread villous atrophy and severe symptoms and co-morbidities. To search for novel CeD biomarkers, we used single-cell RNA sequencing (scRNAseq) to investigate PBMC samples from 11 children before and after seroconversion for CeD and 10 control individuals matched for age, sex and HLA-genotype. We generated scRNAseq profiles of 9559 cells and identified the expected major cellular lineages. Cell proportions remained stable across the different timepoints and health conditions, but we observed differences in gene expression profiles in specific cell types when comparing patient samples before and after disease development and comparing patients with controls. Based on the time when transcripts were differentially expressed, we could classify the deregulated genes as biomarkers for active CeD or as potential pre-diagnostic markers. Pathway analysis showed that active CeD biomarkers display a transcriptional profile associated with antigen activation in CD4+ T cells, whereas NK cells express a subset of biomarker genes even before CeD diagnosis. Intersection of biomarker genes with CeD-associated genetic risk loci pinpointed genetic factors that might play a role in CeD onset. Investigation of potential cellular interaction pathways of PBMC cell subpopulations highlighted the importance of TNF pathways in CeD. Altogether, our results pinpoint genes and pathways that are altered prior to and during CeD onset, thereby identifying novel potential biomarkers for CeD diagnosis in blood.


Asunto(s)
Enfermedad Celíaca , Atrofia , Biomarcadores , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/genética , Niño , Humanos , Leucocitos Mononucleares/metabolismo , Análisis de Secuencia de ARN , Seroconversión
5.
Genome Biol ; 22(1): 198, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34229738

RESUMEN

BACKGROUND: Recent studies highlight the role of metabolites in immune diseases, but it remains unknown how much of this effect is driven by genetic and non-genetic host factors. RESULT: We systematically investigate circulating metabolites in a cohort of 500 healthy subjects (500FG) in whom immune function and activity are deeply measured and whose genetics are profiled. Our data reveal that several major metabolic pathways, including the alanine/glutamate pathway and the arachidonic acid pathway, have a strong impact on cytokine production in response to ex vivo stimulation. We also examine the genetic regulation of metabolites associated with immune phenotypes through genome-wide association analysis and identify 29 significant loci, including eight novel independent loci. Of these, one locus (rs174584-FADS2) associated with arachidonic acid metabolism is causally associated with Crohn's disease, suggesting it is a potential therapeutic target. CONCLUSION: This study provides a comprehensive map of the integration between the blood metabolome and immune phenotypes, reveals novel genetic factors that regulate blood metabolite concentrations, and proposes an integrative approach for identifying new disease treatment targets.


Asunto(s)
Inmunidad Innata/genética , Redes y Vías Metabólicas/genética , Fenotipo , Sitios de Carácter Cuantitativo , Adolescente , Adulto , Anciano , Alanina/sangre , Alanina/inmunología , Ácido Araquidónico/sangre , Ácido Araquidónico/inmunología , Estudios de Cohortes , Femenino , Estudio de Asociación del Genoma Completo , Genómica/métodos , Ácido Glutámico/sangre , Ácido Glutámico/inmunología , Voluntarios Sanos , Humanos , Masculino , Redes y Vías Metabólicas/inmunología , Metabolómica/métodos , Persona de Mediana Edad
6.
Sci Rep ; 11(1): 9252, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33927210

RESUMEN

Celiac disease is an auto-immune disease in which an immune response to dietary gluten leads to inflammation and subsequent atrophy of small intestinal villi, causing severe bowel discomfort and malabsorption of nutrients. The major instigating factor for the immune response in celiac disease is the activation of gluten-specific CD4+ T cells expressing T cell receptors that recognize gluten peptides presented in the context of HLA-DQ2 and DQ8. Here we provide an in-depth characterization of 28 gluten-specific T cell clones. We assess their transcriptional and epigenetic response to T cell receptor stimulation and link this to genetic factors associated with celiac disease. Gluten-specific T cells have a distinct transcriptional profile that mostly resembles that of Th1 cells but also express cytokines characteristic of other types of T-helper cells. This transcriptional response appears not to be regulated by changes in chromatin state, but rather by early upregulation of transcription factors and non-coding RNAs that likely orchestrate the subsequent activation of genes that play a role in immune pathways. Finally, integration of chromatin and transcription factor binding profiles suggest that genes activated by T cell receptor stimulation of gluten­specific T cells may be impacted by genetic variation at several genetic loci associated with celiac disease.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Celíaca/inducido químicamente , Enfermedad Celíaca/patología , Citocinas/inmunología , Citocinas/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glútenes/administración & dosificación , Glútenes/inmunología , Humanos , Receptores de Antígenos de Linfocitos T/genética , Transcriptoma
7.
J Autoimmun ; 108: 102422, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32033836

RESUMEN

The respective effects of tissue alarmins interleukin (IL)-15 and interferon beta (IFNß), and IL-21 produced by T cells on the reprogramming of cytotoxic T lymphocytes (CTLs) that cause tissue destruction in celiac disease is poorly understood. Transcriptomic and epigenetic profiling of primary intestinal CTLs showed massive and distinct temporal transcriptional changes in response to tissue alarmins, while the impact of IL-21 was limited. Only anti-viral pathways were induced in response to all the three stimuli, albeit with differences in dynamics and strength. Moreover, changes in gene expression were primarily independent of changes in H3K27ac, suggesting that other regulatory mechanisms drive the robust transcriptional response. Finally, we found that IL-15/IFNß/IL-21 transcriptional signatures could be linked to transcriptional alterations in risk loci for complex immune diseases. Together these results provide new insights into molecular mechanisms that fuel the activation of CTLs under conditions that emulate the inflammatory environment in patients with autoimmune diseases.


Asunto(s)
Alarminas/metabolismo , Citocinas/metabolismo , Regulación de la Expresión Génica , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Autoinmunidad , Enfermedad Celíaca/etiología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/patología , Perfilación de la Expresión Génica , Humanos , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Interleucina-15/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Regiones Promotoras Genéticas
8.
Hum Mol Genet ; 26(R2): R185-R192, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28977443

RESUMEN

Autoimmune diseases such as rheumatoid arthritis and coeliac disease are typical examples of complex genetic diseases caused by a combination of genetic and non-genetic risk factors. Insight into the genetic risk factors (single nucleotide polymorphisms (SNPs)) has increased since genome-wide association studies (GWAS) became possible in 2007 and, for individual diseases, SNPs can now explain some 15-50% of genetic risk. GWAS have also shown that some 50% of the genetic risk factors for individual autoimmune diseases overlap between different diseases. Thus, shared risk factors may converge to pathways that, when perturbed by genetic variation, predispose to autoimmunity in general. This raises the question of what determines disease specificity, and suggests that identical risk factors may have different effects in various autoimmune diseases. Addressing this question requires translation of genetic risk factors to causal genes and then to molecular and cellular pathways. Since >90% of the genetic risk factors are found in the non-coding part of the genome (i.e. outside the exons of protein-coding genes) and can have an impact on gene regulation, there is an urgent need to better understand the non-coding part of the genome. Here, we will outline the methods being used to unravel the gene regulatory networks perturbed in autoimmune diseases and the importance of doing this in the relevant cell types. We will highlight findings in coeliac disease, which manifests in the small intestine, to demonstrate how cell type and disease context can impact on the consequences of genetic risk factors.


Asunto(s)
Enfermedades Autoinmunes/genética , Redes Reguladoras de Genes/inmunología , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/etiología , Autoinmunidad/genética , Autoinmunidad/inmunología , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Predisposición Genética a la Enfermedad , Variación Genética/genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo , Factores de Riesgo
9.
Nat Protoc ; 11(8): 1455-76, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27442863

RESUMEN

We provide a protocol for precision nuclear run-on sequencing (PRO-seq) and its variant, PRO-cap, which map the location of active RNA polymerases (PRO-seq) or transcription start sites (TSSs) (PRO-cap) genome-wide at high resolution. The density of RNA polymerases at a particular genomic locus directly reflects the level of nascent transcription at that region. Nuclei are isolated from cells and, under nuclear run-on conditions, transcriptionally engaged RNA polymerases incorporate one or, at most, a few biotin-labeled nucleotide triphosphates (biotin-NTPs) into the 3' end of nascent RNA. The biotin-labeled nascent RNA is used to prepare sequencing libraries, which are sequenced from the 3' end to provide high-resolution positional information for the RNA polymerases. PRO-seq provides much higher sensitivity than ChIP-seq, and it generates a much larger fraction of usable sequence reads than ChIP-seq or NET-seq (native elongating transcript sequencing). Similarly to NET-seq, PRO-seq maps the RNA polymerase at up to base-pair resolution with strand specificity, but unlike NET-seq it does not require immunoprecipitation. With the protocol provided here, PRO-seq (or PRO-cap) libraries for high-throughput sequencing can be generated in 4-5 working days. The method has been applied to human, mouse, Drosophila melanogaster and Caenorhabditis elegans cells and, with slight modifications, to yeast.


Asunto(s)
Emparejamiento Base , Mapeo Cromosómico/métodos , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN/química , ARN/genética , Animales , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Humanos , Ratones , ARN/metabolismo , Análisis de Secuencia de ARN , Sitio de Iniciación de la Transcripción
10.
J Autoimmun ; 68: 62-74, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26898941

RESUMEN

Genome-wide association and fine-mapping studies in 14 autoimmune diseases (AID) have implicated more than 250 loci in one or more of these diseases. As more than 90% of AID-associated SNPs are intergenic or intronic, pinpointing the causal genes is challenging. We performed a systematic analysis to link 460 SNPs that are associated with 14 AID to causal genes using transcriptomic data from 629 blood samples. We were able to link 71 (39%) of the AID-SNPs to two or more nearby genes, providing evidence that for part of the AID loci multiple causal genes exist. While 54 of the AID loci are shared by one or more AID, 17% of them do not share candidate causal genes. In addition to finding novel genes such as ULK3, we also implicate novel disease mechanisms and pathways like autophagy in celiac disease pathogenesis. Furthermore, 42 of the AID SNPs specifically affected the expression of 53 non-coding RNA genes. To further understand how the non-coding genome contributes to AID, the SNPs were linked to functional regulatory elements, which suggest a model where AID genes are regulated by network of chromatin looping/non-coding RNAs interactions. The looping model also explains how a causal candidate gene is not necessarily the gene closest to the AID SNP, which was the case in nearly 50% of cases.


Asunto(s)
Enfermedades Autoinmunes/genética , Mapeo Cromosómico , Expresión Génica , Variación Genética , Estudio de Asociación del Genoma Completo , Sitios de Carácter Cuantitativo , ARN no Traducido , Enfermedades Autoinmunes/metabolismo , Autofagia/genética , Enfermedad Celíaca/genética , Enfermedad Celíaca/metabolismo , Citocinas/metabolismo , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Genoma Humano , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Desequilibrio de Ligamiento , Polimorfismo de Nucleótido Simple , ARN Largo no Codificante/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...