Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brief Bioinform ; 24(5)2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37587790

RESUMO

Precision medicine relies on the identification of robust disease and risk factor signatures from omics data. However, current knowledge-driven approaches may overlook novel or unexpected phenomena due to the inherent biases in biological knowledge. In this study, we present a data-driven signature discovery workflow for DNA methylation analysis utilizing network-coherent autoencoders (NCAEs) with biologically relevant latent embeddings. First, we explored the architecture space of autoencoders trained on a large-scale pan-tissue compendium (n = 75 272) of human epigenome-wide association studies. We observed the emergence of co-localized patterns in the deep autoencoder latent space representations that corresponded to biological network modules. We determined the NCAE configuration with the strongest co-localization and centrality signals in the human protein interactome. Leveraging the NCAE embeddings, we then trained interpretable deep neural networks for risk factor (aging, smoking) and disease (systemic lupus erythematosus) prediction and classification tasks. Remarkably, our NCAE embedding-based models outperformed existing predictors, revealing novel DNA methylation signatures enriched in gene sets and pathways associated with the studied condition in each case. Our data-driven biomarker discovery workflow provides a generally applicable pipeline to capture relevant risk factor and disease information. By surpassing the limitations of knowledge-driven methods, our approach enhances the understanding of complex epigenetic processes, facilitating the development of more effective diagnostic and therapeutic strategies.


Assuntos
Algoritmos , Metilação de DNA , Humanos , Redes Neurais de Computação , Epigênese Genética , Fatores de Risco
2.
Proc Natl Acad Sci U S A ; 120(14): e2212476120, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-36989306

RESUMO

Endothelial dysfunction and impaired vasodilation are linked with adverse cardiovascular events. T lymphocytes expressing choline acetyltransferase (ChAT), the enzyme catalyzing biosynthesis of the vasorelaxant acetylcholine (ACh), regulate vasodilation and are integral to the cholinergic antiinflammatory pathway in an inflammatory reflex in mice. Here, we found that human T cell ChAT mRNA expression was induced by T cell activation involving the PI3K signaling cascade. Mechanistically, we identified that ChAT mRNA expression was induced following the attenuation of RE-1 Silencing Transcription factor REST-mediated methylation of the ChAT promoter, and that ChAT mRNA expression levels were up-regulated by GATA3 in human T cells. In functional experiments, T cell-derived ACh increased endothelial nitric oxide-synthase activity, promoted vasorelaxation, and reduced vascular endothelial activation and promoted barrier integrity by a cholinergic mechanism. Further, we observed that survival in a cohort of patients with severe circulatory failure correlated with their relative frequency of ChAT +CD4+ T cells in blood. These findings on ChAT+ human T cells provide a mechanism for cholinergic immune regulation of vascular endothelial function in human inflammation.


Assuntos
Colina O-Acetiltransferase , Linfócitos T , Humanos , Camundongos , Animais , Linfócitos T/metabolismo , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Colinérgicos , Acetilcolina/metabolismo , RNA Mensageiro/metabolismo
3.
BMC Genomics ; 22(1): 631, 2021 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-34461822

RESUMO

BACKGROUND: There exist few, if any, practical guidelines for predictive and falsifiable multi-omic data integration that systematically integrate existing knowledge. Disease modules are popular concepts for interpreting genome-wide studies in medicine but have so far not been systematically evaluated and may lead to corroborating multi-omic modules. RESULT: We assessed eight module identification methods in 57 previously published expression and methylation studies of 19 diseases using GWAS enrichment analysis. Next, we applied the same strategy for multi-omic integration of 20 datasets of multiple sclerosis (MS), and further validated the resulting module using both GWAS and risk-factor-associated genes from several independent cohorts. Our benchmark of modules showed that in immune-associated diseases modules inferred from clique-based methods were the most enriched for GWAS genes. The multi-omic case study using MS data revealed the robust identification of a module of 220 genes. Strikingly, most genes of the module were differentially methylated upon the action of one or several environmental risk factors in MS (n = 217, P = 10- 47) and were also independently validated for association with five different risk factors of MS, which further stressed the high genetic and epigenetic relevance of the module for MS. CONCLUSIONS: We believe our analysis provides a workflow for selecting modules and our benchmark study may help further improvement of disease module methods. Moreover, we also stress that our methodology is generally applicable for combining and assessing the performance of multi-omic approaches for complex diseases.


Assuntos
Estudo de Associação Genômica Ampla , Esclerose Múltipla , Epigenômica , Redes Reguladoras de Genes , Humanos , Esclerose Múltipla/genética , Fatores de Risco
4.
Clin Epigenetics ; 13(1): 135, 2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34193262

RESUMO

BACKGROUND: Environmental exposures may alter DNA methylation patterns of T helper cells. As T helper cells are instrumental for allergy development, changes in methylation patterns may constitute a mechanism of action for allergy preventive interventions. While epigenetic effects of separate perinatal probiotic or ω-3 fatty acid supplementation have been studied previously, the combined treatment has not been assessed. We aimed to investigate epigenome-wide DNA methylation patterns from a sub-group of children in an on-going randomised double-blind placebo-controlled allergy prevention trial using pre- and postnatal combined Lactobacillus reuteri and ω-3 fatty acid treatment. To this end, > 866000 CpG sites (MethylationEPIC 850K array) in cord blood CD4+ T cells were examined in samples from all four study arms (double-treatment: n = 18, single treatments: probiotics n = 16, ω-3 n = 15, and double placebo: n = 14). Statistical and bioinformatic analyses identified treatment-associated differentially methylated CpGs and genes, which were used to identify putatively treatment-induced network modules. Pathway analyses inferred biological relevance, and comparisons were made to an independent allergy data set. RESULTS: Comparing the active treatments to the double placebo group, most differentially methylated CpGs and genes were hypermethylated, possibly suggesting induction of transcriptional inhibition. The double-treated group showed the largest number of differentially methylated CpGs, of which many were unique, suggesting synergy between interventions. Clusters within the double-treated network module consisted of immune-related pathways, including T cell receptor signalling, and antigen processing and presentation, with similar pathways revealed for the single-treatment modules. CpGs derived from differential methylation and network module analyses were enriched in an independent allergy data set, particularly in the double-treatment group, proposing treatment-induced DNA methylation changes as relevant for allergy development. CONCLUSION: Prenatal L. reuteri and/or ω-3 fatty acid treatment results in hypermethylation and affects immune- and allergy-related pathways in neonatal T helper cells, with potentially synergistic effects between the interventions and relevance for allergic disease. Further studies need to address these findings on a transcriptional level, and whether the results associate to allergy development in the children. Understanding the role of DNA methylation in regulating effects of perinatal probiotic and ω-3 interventions may provide essential knowledge in the development of efficacious allergy preventive strategies. Trial registration ClinicalTrials.gov, ClinicalTrials.gov-ID: NCT01542970. Registered 27th of February 2012-Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT01542970 .


Assuntos
Metilação de DNA/efeitos dos fármacos , Ácidos Graxos Ômega-3/metabolismo , Limosilactobacillus reuteri/metabolismo , Adulto , Suplementos Nutricionais/normas , Ácidos Graxos Ômega-3/administração & dosagem , Feminino , Humanos , Saúde do Lactente , Recém-Nascido , Limosilactobacillus reuteri/patogenicidade , Masculino , Placebos , Gravidez , Cuidado Pré-Natal/métodos , Cuidado Pré-Natal/tendências
6.
Bioinformatics ; 36(12): 3918-3919, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32271876

RESUMO

MOTIVATION: Complex diseases are due to the dense interactions of many disease-associated factors that dysregulate genes that in turn form the so-called disease modules, which have shown to be a powerful concept for understanding pathological mechanisms. There exist many disease module inference methods that rely on somewhat different assumptions, but there is still no gold standard or best-performing method. Hence, there is a need for combining these methods to generate robust disease modules. RESULTS: We developed MODule IdentiFIER (MODifieR), an ensemble R package of nine disease module inference methods from transcriptomics networks. MODifieR uses standardized input and output allowing the possibility to combine individual modules generated from these methods into more robust disease-specific modules, contributing to a better understanding of complex diseases. AVAILABILITY AND IMPLEMENTATION: MODifieR is available under the GNU GPL license and can be freely downloaded from https://gitlab.com/Gustafsson-lab/MODifieR and as a Docker image from https://hub.docker.com/r/ddeweerd/modifier. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Biologia Computacional , Software , Transcriptoma
7.
Genome Med ; 11(1): 47, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31358043

RESUMO

BACKGROUND: Genomic medicine has paved the way for identifying biomarkers and therapeutically actionable targets for complex diseases, but is complicated by the involvement of thousands of variably expressed genes across multiple cell types. Single-cell RNA-sequencing study (scRNA-seq) allows the characterization of such complex changes in whole organs. METHODS: The study is based on applying network tools to organize and analyze scRNA-seq data from a mouse model of arthritis and human rheumatoid arthritis, in order to find diagnostic biomarkers and therapeutic targets. Diagnostic validation studies were performed using expression profiling data and potential protein biomarkers from prospective clinical studies of 13 diseases. A candidate drug was examined by a treatment study of a mouse model of arthritis, using phenotypic, immunohistochemical, and cellular analyses as read-outs. RESULTS: We performed the first systematic analysis of pathways, potential biomarkers, and drug targets in scRNA-seq data from a complex disease, starting with inflamed joints and lymph nodes from a mouse model of arthritis. We found the involvement of hundreds of pathways, biomarkers, and drug targets that differed greatly between cell types. Analyses of scRNA-seq and GWAS data from human rheumatoid arthritis (RA) supported a similar dispersion of pathogenic mechanisms in different cell types. Thus, systems-level approaches to prioritize biomarkers and drugs are needed. Here, we present a prioritization strategy that is based on constructing network models of disease-associated cell types and interactions using scRNA-seq data from our mouse model of arthritis, as well as human RA, which we term multicellular disease models (MCDMs). We find that the network centrality of MCDM cell types correlates with the enrichment of genes harboring genetic variants associated with RA and thus could potentially be used to prioritize cell types and genes for diagnostics and therapeutics. We validated this hypothesis in a large-scale study of patients with 13 different autoimmune, allergic, infectious, malignant, endocrine, metabolic, and cardiovascular diseases, as well as a therapeutic study of the mouse arthritis model. CONCLUSIONS: Overall, our results support that our strategy has the potential to help prioritize diagnostic and therapeutic targets in human disease.


Assuntos
Suscetibilidade a Doenças , Técnicas de Diagnóstico Molecular , Herança Multifatorial , Análise de Célula Única , Animais , Artrite Reumatoide/diagnóstico , Artrite Reumatoide/etiologia , Biomarcadores , Biologia Computacional/métodos , Modelos Animais de Doenças , Descoberta de Drogas/métodos , Perfilação da Expressão Gênica , Genômica/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Redes Neurais de Computação , Reprodutibilidade dos Testes , Análise de Célula Única/métodos
8.
Genome Med ; 12(1): 4, 2019 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-31892363

RESUMO

Personalized medicine requires the integration and processing of vast amounts of data. Here, we propose a solution to this challenge that is based on constructing Digital Twins. These are high-resolution models of individual patients that are computationally treated with thousands of drugs to find the drug that is optimal for the patient.


Assuntos
Medicina de Precisão , Bases de Dados Factuais , Doença/genética , Humanos , Redes Neurais de Computação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...