Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Genome Med ; 16(1): 42, 2024 03 20.
Article in English | MEDLINE | ID: mdl-38509600

ABSTRACT

BACKGROUND: Ineffective drug treatment is a major problem for many patients with immune-mediated inflammatory diseases (IMIDs). Important reasons are the lack of systematic solutions for drug prioritisation and repurposing based on characterisation of the complex and heterogeneous cellular and molecular changes in IMIDs. METHODS: Here, we propose a computational framework, scDrugPrio, which constructs network models of inflammatory disease based on single-cell RNA sequencing (scRNA-seq) data. scDrugPrio constructs detailed network models of inflammatory diseases that integrate information on cell type-specific expression changes, altered cellular crosstalk and pharmacological properties for the selection and ranking of thousands of drugs. RESULTS: scDrugPrio was developed using a mouse model of antigen-induced arthritis and validated by improved precision/recall for approved drugs, as well as extensive in vitro, in vivo, and in silico studies of drugs that were predicted, but not approved, for the studied diseases. Next, scDrugPrio was applied to multiple sclerosis, Crohn's disease, and psoriatic arthritis, further supporting scDrugPrio through prioritisation of relevant and approved drugs. However, in contrast to the mouse model of arthritis, great interindividual cellular and gene expression differences were found in patients with the same diagnosis. Such differences could explain why some patients did or did not respond to treatment. This explanation was supported by the application of scDrugPrio to scRNA-seq data from eleven individual Crohn's disease patients. The analysis showed great variations in drug predictions between patients, for example, assigning a high rank to anti-TNF treatment in a responder and a low rank in a nonresponder to that treatment. CONCLUSIONS: We propose a computational framework, scDrugPrio, for drug prioritisation based on scRNA-seq of IMID disease. Application to individual patients indicates scDrugPrio's potential for personalised network-based drug screening on cellulome-, genome-, and drugome-wide scales. For this purpose, we made scDrugPrio into an easy-to-use R package ( https://github.com/SDTC-CPMed/scDrugPrio ).


Subject(s)
Arthritis , Crohn Disease , Humans , Precision Medicine , Tumor Necrosis Factor Inhibitors , Gene Expression Profiling , Immunomodulating Agents , Single-Cell Analysis , Sequence Analysis, RNA
2.
bioRxiv ; 2023 Nov 13.
Article in English | MEDLINE | ID: mdl-38014022

ABSTRACT

Background: Ineffective drug treatment is a major problem for many patients with immune-mediated inflammatory diseases (IMIDs). Important reasons are the lack of systematic solutions for drug prioritisation and repurposing based on characterisation of the complex and heterogeneous cellular and molecular changes in IMIDs. Methods: Here, we propose a computational framework, scDrugPrio, which constructs network models of inflammatory disease based on single-cell RNA sequencing (scRNA-seq) data. scDrugPrio constructs detailed network models of inflammatory diseases that integrate information on cell type-specific expression changes, altered cellular crosstalk and pharmacological properties for the selection and ranking of thousands of drugs. Results: scDrugPrio was developed using a mouse model of antigen-induced arthritis and validated by improved precision/recall for approved drugs, as well as extensive in vitro, in vivo, and in silico studies of drugs that were predicted, but not approved, for the studied diseases. Next, scDrugPrio was applied to multiple sclerosis, Crohn's disease, and psoriatic arthritis, further supporting scDrugPrio through prioritisation of relevant and approved drugs. However, in contrast to the mouse model of arthritis, great interindividual cellular and gene expression differences were found in patients with the same diagnosis. Such differences could explain why some patients did or did not respond to treatment. This explanation was supported by the application of scDrugPrio to scRNA-seq data from eleven individual Crohn's disease patients. The analysis showed great variations in drug predictions between patients, for example, assigning a high rank to anti-TNF treatment in a responder and a low rank in a nonresponder to that treatment. Conclusion: We propose a computational framework, scDrugPrio, for drug prioritisation based on scRNA-seq of IMID disease. Application to individual patients indicates scDrugPrio's potential for personalised network-based drug screening on cellulome-, genome-, and drugome-wide scales. For this purpose, we made scDrugPrio into an easy-to-use R package (https://github.com/SDTC-CPMed/scDrugPrio).

3.
Cell Rep Med ; 4(3): 100956, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36858042

ABSTRACT

Prioritization of disease mechanisms, biomarkers, and drug targets in immune-mediated inflammatory diseases (IMIDs) is complicated by altered interactions between thousands of genes. Our multi-organ single-cell RNA sequencing of a mouse IMID model, namely collagen-induced arthritis, shows highly complex and heterogeneous expression changes in all analyzed organs, even though only joints showed signs of inflammation. We organized those into a multi-organ multicellular disease model, which shows predicted molecular interactions within and between organs. That model supports that inflammation is switched on or off by altered balance between pro- and anti-inflammatory upstream regulators (URs) and downstream pathways. Meta-analyses of human IMIDs show a similar, but graded, on/off switch system. This system has the potential to prioritize, diagnose, and treat optimal combinations of URs on the levels of IMIDs, subgroups, and individual patients. That potential is supported by UR analyses in more than 600 sera from patients with systemic lupus erythematosus.


Subject(s)
Immune System Diseases , Immunomodulating Agents , Animals , Mice , Humans , Precision Medicine , Inflammation/metabolism , Immune System Diseases/genetics , Immune System Diseases/therapy , Single-Cell Analysis
4.
Genome Med ; 14(1): 48, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35513850

ABSTRACT

BACKGROUND: Medical digital twins are computational disease models for drug discovery and treatment. Unresolved problems include how to organize and prioritize between disease-associated changes in digital twins, on cellulome- and genome-wide scales. We present a dynamic framework that can be used to model such changes and thereby prioritize upstream regulators (URs) for biomarker- and drug discovery. METHODS: We started with seasonal allergic rhinitis (SAR) as a disease model, by analyses of in vitro allergen-stimulated peripheral blood mononuclear cells (PBMC) from SAR patients. Time-series a single-cell RNA-sequencing (scRNA-seq) data of these cells were used to construct multicellular network models (MNMs) at each time point of molecular interactions between cell types. We hypothesized that predicted molecular interactions between cell types in the MNMs could be traced to find an UR gene, at an early time point. We performed bioinformatic and functional studies of the MNMs to develop a scalable framework to prioritize UR genes. This framework was tested on a single-cell and bulk-profiling data from SAR and other inflammatory diseases. RESULTS: Our scRNA-seq-based time-series MNMs of SAR showed thousands of differentially expressed genes (DEGs) across multiple cell types, which varied between time points. Instead of a single-UR gene in each MNM, we found multiple URs dispersed across the cell types. Thus, at each time point, the MNMs formed multi-directional networks. The absence of linear hierarchies and time-dependent variations in MNMs complicated the prioritization of URs. For example, the expression and functions of Th2 cytokines, which are approved drug targets in allergies, varied across cell types, and time points. Our analyses of bulk- and single-cell data from other inflammatory diseases also revealed multi-directional networks that showed stage-dependent variations. We therefore developed a quantitative approach to prioritize URs: we ranked the URs based on their predicted effects on downstream target cells. Experimental and bioinformatic analyses supported that this kind of ranking is a tractable approach for prioritizing URs. CONCLUSIONS: We present a scalable framework for modeling dynamic changes in digital twins, on cellulome- and genome-wide scales, to prioritize UR genes for biomarker and drug discovery.


Subject(s)
Gene Expression Profiling , Single-Cell Analysis , Biomarkers/metabolism , Computational Biology , Humans , Leukocytes, Mononuclear/metabolism
5.
Lakartidningen ; 1182021 05 11.
Article in Swedish | MEDLINE | ID: mdl-33977515

ABSTRACT

Recent technical developments and early clinical examples support that precision medicine has potential to provide novel diagnostic and therapeutic solutions for patients with complex diseases, who are not responding to existing therapies. Those solutions will require integration of genomic data with routine clinical, imaging, sensor, biobank and registry data. Moreover, user-friendly tools for informed decision support for both patients and clinicians will be needed. While this will entail huge technical, ethical, societal and regulatory challenges, it may contribute to transforming and improving health care towards becoming predictive, preventive, personalised and participatory (4P-medicine).


Subject(s)
Genomics , Precision Medicine , Delivery of Health Care , Humans
6.
J Immunol Res ; 2020: 8279619, 2020.
Article in English | MEDLINE | ID: mdl-32411805

ABSTRACT

BACKGROUND: Unbiased studies using different genome-wide methods have identified a great number of candidate biomarkers for diagnosis and treatment response in pediatric ulcerative colitis (UC). However, clinical translation has been proven difficult. Here, we hypothesized that one reason could be differences between inflammatory responses in an inflamed gut and in peripheral blood cells. METHODS: We performed meta-analysis of gene expression microarray data from intestinal biopsies and whole blood cells (WBC) from pediatric patients with UC and healthy controls in order to identify overlapping pathways, predicted upstream regulators, and potential biomarkers. RESULTS: Analyses of profiling datasets from colonic biopsies showed good agreement between different studies regarding pathways and predicted upstream regulators. The most activated predicted upstream regulators included TNF, which is known to have a key pathogenic and therapeutic role in pediatric UC. Despite this, the expression levels of TNF were increased in neither colonic biopsies nor WBC. A potential explanation was increased expression of TNFR2, one of the membrane-bound receptors of TNF in the inflamed colon. Further analyses showed a similar pattern of complex relations between the expression levels of the regulators and their receptors. We also found limited overlap between pathways and predicted upstream regulators in colonic biopsies and WBC. An extended search including all differentially expressed genes that overlapped between colonic biopsies and WBC only resulted in identification of three potential biomarkers involved in the regulation of intestinal inflammation. However, two had been previously proposed in adult inflammatory bowel diseases (IBD), namely, MMP9 and PROK2. CONCLUSIONS: Our findings indicate that biomarker identification in pediatric UC is complicated by the involvement of multiple pathways, each of which includes many different types of genes in the blood or inflamed intestine. Therefore, further studies for identification of combinatorial biomarkers are warranted. Our study may provide candidate biomarkers for such studies.


Subject(s)
Colitis, Ulcerative/diagnosis , Colon/pathology , Intestinal Mucosa/pathology , Biomarkers/analysis , Biopsy , Child , Colitis, Ulcerative/blood , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Colon/immunology , Gastrointestinal Hormones/analysis , Gastrointestinal Hormones/genetics , Gene Expression Profiling , Humans , Intestinal Mucosa/immunology , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinase 9/genetics , Neuropeptides/analysis , Neuropeptides/genetics , Oligonucleotide Array Sequence Analysis , Receptors, Tumor Necrosis Factor, Type II/analysis , Receptors, Tumor Necrosis Factor, Type II/genetics , Treatment Outcome
8.
Sci Rep ; 9(1): 15575, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666584

ABSTRACT

Screening programs for colorectal cancer (CRC) often rely on detection of blood in stools, which is unspecific and leads to a large number of colonoscopies of healthy subjects. Painstaking research has led to the identification of a large number of different types of biomarkers, few of which are in general clinical use. Here, we searched for highly accurate combinations of biomarkers by meta-analyses of genome- and proteome-wide data from CRC tumors. We focused on secreted proteins identified by the Human Protein Atlas and used our recently described algorithms to find optimal combinations of proteins. We identified nine proteins, three of which had been previously identified as potential biomarkers for CRC, namely CEACAM5, LCN2 and TRIM28. The remaining proteins were PLOD1, MAD1L1, P4HA1, GNS, C12orf10 and P3H1. We analyzed these proteins in plasma from 80 patients with newly diagnosed CRC and 80 healthy controls. A combination of four of these proteins, TRIM28, PLOD1, CEACAM5 and P4HA1, separated a training set consisting of 90% patients and 90% of the controls with high accuracy, which was verified in a test set consisting of the remaining 10%. Further studies are warranted to test our algorithms and proteins for early CRC diagnosis.


Subject(s)
Algorithms , Biomarkers, Tumor/blood , Colorectal Neoplasms/blood , Proteomics/methods , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Humans
9.
Genome Med ; 11(1): 47, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31358043

ABSTRACT

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.


Subject(s)
Disease Susceptibility , Molecular Diagnostic Techniques , Multifactorial Inheritance , Single-Cell Analysis , Animals , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/etiology , Biomarkers , Computational Biology/methods , Disease Models, Animal , Drug Discovery/methods , Gene Expression Profiling , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Mice , Neural Networks, Computer , Reproducibility of Results , Single-Cell Analysis/methods
10.
Genome Med ; 12(1): 4, 2019 12 31.
Article in English | MEDLINE | ID: mdl-31892363

ABSTRACT

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.


Subject(s)
Precision Medicine , Databases, Factual , Disease/genetics , Humans , Neural Networks, Computer
11.
PLoS Comput Biol ; 13(6): e1005608, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28640810

ABSTRACT

Recent technological advancements have made time-resolved, quantitative, multi-omics data available for many model systems, which could be integrated for systems pharmacokinetic use. Here, we present large-scale simulation modeling (LASSIM), which is a novel mathematical tool for performing large-scale inference using mechanistically defined ordinary differential equations (ODE) for gene regulatory networks (GRNs). LASSIM integrates structural knowledge about regulatory interactions and non-linear equations with multiple steady state and dynamic response expression datasets. The rationale behind LASSIM is that biological GRNs can be simplified using a limited subset of core genes that are assumed to regulate all other gene transcription events in the network. The LASSIM method is implemented as a general-purpose toolbox using the PyGMO Python package to make the most of multicore computers and high performance clusters, and is available at https://gitlab.com/Gustafsson-lab/lassim. As a method, LASSIM works in two steps, where it first infers a non-linear ODE system of the pre-specified core gene expression. Second, LASSIM in parallel optimizes the parameters that model the regulation of peripheral genes by core system genes. We showed the usefulness of this method by applying LASSIM to infer a large-scale non-linear model of naïve Th2 cell differentiation, made possible by integrating Th2 specific bindings, time-series together with six public and six novel siRNA-mediated knock-down experiments. ChIP-seq showed significant overlap for all tested transcription factors. Next, we performed novel time-series measurements of total T-cells during differentiation towards Th2 and verified that our LASSIM model could monitor those data significantly better than comparable models that used the same Th2 bindings. In summary, the LASSIM toolbox opens the door to a new type of model-based data analysis that combines the strengths of reliable mechanistic models with truly systems-level data. We demonstrate the power of this approach by inferring a mechanistically motivated, genome-wide model of the Th2 transcription regulatory system, which plays an important role in several immune related diseases.


Subject(s)
Chromosome Mapping/methods , Models, Genetic , Proteome/metabolism , Signal Transduction/physiology , Software , Th2 Cells/metabolism , Algorithms , Cell Differentiation/physiology , Cells, Cultured , Computer Simulation , Gene Expression Regulation, Developmental/physiology , Humans , Programming Languages
12.
Cell Rep ; 16(11): 2928-2939, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27626663

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS and has a varying disease course as well as variable response to treatment. Biomarkers may therefore aid personalized treatment. We tested whether in vitro activation of MS patient-derived CD4+ T cells could reveal potential biomarkers. The dynamic gene expression response to activation was dysregulated in patient-derived CD4+ T cells. By integrating our findings with genome-wide association studies, we constructed a highly connected MS gene module, disclosing cell activation and chemotaxis as central components. Changes in several module genes were associated with differences in protein levels, which were measurable in cerebrospinal fluid and were used to classify patients from control individuals. In addition, these measurements could predict disease activity after 2 years and distinguish low and high responders to treatment in two additional, independent cohorts. While further validation is needed in larger cohorts prior to clinical implementation, we have uncovered a set of potentially promising biomarkers.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Protein Interaction Maps/genetics , Adult , Case-Control Studies , Cerebrospinal Fluid Proteins/metabolism , Chemotaxis/genetics , Cohort Studies , Female , Gene Expression Profiling , Genome-Wide Association Study , Humans , Lymphocyte Activation/genetics , Male , Middle Aged , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/pathology , Prognosis , Young Adult
13.
Int J Endocrinol ; 2016: 1427042, 2016.
Article in English | MEDLINE | ID: mdl-27656207

ABSTRACT

Papillary thyroid cancer (PTC) can be divided into classical variant of PTC (cPTC), follicular variant of PTC (fvPTC), and tall cell variant (tcPTC). These variants differ in their histopathology and cytology; however, their molecular background is not clearly understood. Our results shed some new light on papillary thyroid cancer biology as new direct miRNA-gene regulations are discovered. The Cancer Genome Atlas (TCGA) 466 thyroid cancer samples were studied in parallel datasets to discover potential miRNA-mRNA regulations. Additionally, miRNAs and genes differentiating PTC variants (cPTC, fvPTC, and tcPTC) were indicated. Putative miRNA regulatory pairs were discovered: hsa-miR-146b-5p with PHKB and IRAK1, hsa-miR-874-3p with ITGB4 characteristic for classic PTC samples, and hsa-miR-152-3p with TGFA characteristic for follicular variant PTC samples. MiRNA-mRNA regulations discovery opens a new perspective in understanding of PTC biology. Furthermore, our successful pipeline of miRNA-mRNA regulatory pathways discovery could serve as a universal tool to find new miRNA-mRNA regulations, also in different datasets.

14.
Cell Rep ; 16(2): 559-570, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27346350

ABSTRACT

5-methylcytosine (5mC) is converted to 5-hydroxymethylcytosine (5hmC) by the TET family of enzymes as part of a recently discovered active DNA de-methylation pathway. 5hmC plays important roles in regulation of gene expression and differentiation and has been implicated in T cell malignancies and autoimmunity. Here, we report early and widespread 5mC/5hmC remodeling during human CD4(+) T cell differentiation ex vivo at genes and cell-specific enhancers with known T cell function. We observe similar DNA de-methylation in CD4(+) memory T cells in vivo, indicating that early remodeling events persist long term in differentiated cells. Underscoring their important function, 5hmC loci were highly enriched for genetic variants associated with T cell diseases and T-cell-specific chromosomal interactions. Extensive functional validation of 22 risk variants revealed potentially pathogenic mechanisms in diabetes and multiple sclerosis. Our results support 5hmC-mediated DNA de-methylation as a key component of CD4(+) T cell biology in humans, with important implications for gene regulation and lineage commitment.


Subject(s)
5-Methylcytosine/analogs & derivatives , CD4-Positive T-Lymphocytes/physiology , Cell Differentiation , 5-Methylcytosine/metabolism , Cell Lineage , Cells, Cultured , DNA Methylation , Gene Expression Regulation/immunology , Humans
15.
J Immunol Res ; 2016: 5153184, 2016.
Article in English | MEDLINE | ID: mdl-28097155

ABSTRACT

Specific immunotherapy (SIT) reverses the symptoms of seasonal allergic rhinitis (SAR) in most patients. Recent studies report type I interferons shifting the balance between type I T helper cell (Th1) and type II T helper cells (Th2) towards Th2 dominance by inhibiting the differentiation of naive T cells into Th1 cells. As SIT is thought to cause a shift towards Th1 dominance, we hypothesized that SIT would alter interferon type I signaling. To test this, allergen and diluent challenged CD4+ T cells from healthy controls and patients from different time points were analyzed. The initial experiments focused on signature genes of the pathway and found complex changes following immunotherapy, which were consistent with our hypothesis. As interferon signaling involves multiple genes, expression profiling studies were performed, showing altered expression of the pathway. These findings require validation in a larger group of patients in further studies.


Subject(s)
Immunotherapy/methods , Interferon-alpha/immunology , Interferon-beta/immunology , Rhinitis, Allergic, Seasonal/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Adult , Betula/immunology , Cells, Cultured , Female , Humans , Interferon-alpha/genetics , Interferon-beta/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Leukocytes, Mononuclear/immunology , Middle Aged , Pollen/immunology , Principal Component Analysis , Rhinitis, Allergic, Seasonal/therapy , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/immunology
16.
Sci Transl Med ; 7(313): 313ra178, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26560356

ABSTRACT

Early regulators of disease may increase understanding of disease mechanisms and serve as markers for presymptomatic diagnosis and treatment. However, early regulators are difficult to identify because patients generally present after they are symptomatic. We hypothesized that early regulators of T cell-associated diseases could be found by identifying upstream transcription factors (TFs) in T cell differentiation and by prioritizing hub TFs that were enriched for disease-associated polymorphisms. A gene regulatory network (GRN) was constructed by time series profiling of the transcriptomes and methylomes of human CD4(+) T cells during in vitro differentiation into four helper T cell lineages, in combination with sequence-based TF binding predictions. The TFs GATA3, MAF, and MYB were identified as early regulators and validated by ChIP-seq (chromatin immunoprecipitation sequencing) and small interfering RNA knockdowns. Differential mRNA expression of the TFs and their targets in T cell-associated diseases supports their clinical relevance. To directly test if the TFs were altered early in disease, T cells from patients with two T cell-mediated diseases, multiple sclerosis and seasonal allergic rhinitis, were analyzed. Strikingly, the TFs were differentially expressed during asymptomatic stages of both diseases, whereas their targets showed altered expression during symptomatic stages. This analytical strategy to identify early regulators of disease by combining GRNs with genome-wide association studies may be generally applicable for functional and clinical studies of early disease development.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Regulatory Networks , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Rhinitis, Allergic, Seasonal/genetics , Rhinitis, Allergic, Seasonal/immunology , CD4-Positive T-Lymphocytes/metabolism , GATA3 Transcription Factor/genetics , Genome-Wide Association Study , Humans , Multiple Sclerosis/diagnosis , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-myb/genetics , Rhinitis, Allergic, Seasonal/diagnosis , Transcriptome
17.
Genome Med ; 6(2): 17, 2014.
Article in English | MEDLINE | ID: mdl-24571673

ABSTRACT

BACKGROUND: Translational research typically aims to identify and functionally validate individual, disease-specific genes. However, reaching this aim is complicated by the involvement of thousands of genes in common diseases, and that many of those genes are pleiotropic, that is, shared by several diseases. METHODS: We integrated genomic meta-analyses with prospective clinical studies to systematically investigate the pathogenic, diagnostic and therapeutic roles of pleiotropic genes. In a novel approach, we first used pathway analysis of all published genome-wide association studies (GWAS) to find a cell type common to many diseases. RESULTS: The analysis showed over-representation of the T helper cell differentiation pathway, which is expressed in T cells. This led us to focus on expression profiling of CD4(+) T cells from highly diverse inflammatory and malignant diseases. We found that pleiotropic genes were highly interconnected and formed a pleiotropic module, which was enriched for inflammatory, metabolic and proliferative pathways. The general relevance of this module was supported by highly significant enrichment of genetic variants identified by all GWAS and cancer studies, as well as known diagnostic and therapeutic targets. Prospective clinical studies of multiple sclerosis and allergy showed the importance of both pleiotropic and disease specific modules for clinical stratification. CONCLUSIONS: In summary, this translational genomics study identified a pleiotropic module, which has key pathogenic, diagnostic and therapeutic roles.

18.
Math Biosci Eng ; 10(3): 667-690, 2013 06.
Article in English | MEDLINE | ID: mdl-23906143

ABSTRACT

The problem of feature selection for large-scale genomic data, for example from DNA microarray experiments, is one of the fundamental and well-investigated problems in modern computational biology. From the computational point of view, a selected gene list should be characterized by good predictive power and should be understood and well explained from the biological point of view. Recently, another feature of selected gene lists is increasingly investigated, namely their stability which measures how the content and/or the gene order change when the data are perturbed. In this paper we propose a new approach to analysis of gene list stability, termed the sensitivity index, that does not require any data perturbation and allows the gene list that is most reliable in a biological sense to be chosen.


Subject(s)
Databases, Genetic/statistics & numerical data , Analysis of Variance , Bayes Theorem , Colonic Neoplasms/genetics , Computational Biology , Data Interpretation, Statistical , Female , Humans , Linear Models , Mathematical Concepts , Models, Genetic , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Ovarian Neoplasms/genetics , Population Dynamics , Signal-To-Noise Ratio , Support Vector Machine , Systems Biology
SELECTION OF CITATIONS
SEARCH DETAIL
...