SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "(WFRF:(Gustafsson Mika)) srt2:(2020-2024)"

Sökning: (WFRF:(Gustafsson Mika)) > (2020-2024)

  • Resultat 1-10 av 39
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Kanoni, Stavroula, et al. (författare)
  • Implicating genes, pleiotropy, and sexual dimorphism at blood lipid loci through multi-ancestry meta-analysis.
  • 2022
  • Ingår i: Genome biology. - : Springer Science and Business Media LLC. - 1474-760X .- 1465-6906 .- 1474-7596. ; 23:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Genetic variants within nearly 1000 loci are known to contribute to modulation of blood lipid levels. However, the biological pathways underlying these associations are frequently unknown, limiting understanding of these findings and hindering downstream translational efforts such as drug target discovery.To expand our understanding of the underlying biological pathways and mechanisms controlling blood lipid levels, we leverage a large multi-ancestry meta-analysis (N=1,654,960) of blood lipids to prioritize putative causal genes for 2286 lipid associations using six gene prediction approaches. Using phenome-wide association (PheWAS) scans, we identify relationships of genetically predicted lipid levels to other diseases and conditions. We confirm known pleiotropic associations with cardiovascular phenotypes and determine novel associations, notably with cholelithiasis risk. We perform sex-stratified GWAS meta-analysis of lipid levels and show that 3-5% of autosomal lipid-associated loci demonstrate sex-biased effects. Finally, we report 21 novel lipid loci identified on the X chromosome. Many of the sex-biased autosomal and X chromosome lipid loci show pleiotropic associations with sex hormones, emphasizing the role of hormone regulation in lipid metabolism.Taken together, our findings provide insights into the biological mechanisms through which associated variants lead to altered lipid levels and potentially cardiovascular disease risk.
  •  
2.
  • Badam, Tejaswi, et al. (författare)
  • CD4(+) T-cell DNA methylation changes during pregnancy significantly correlate with disease-associated methylation changes in autoimmune diseases
  • 2022
  • Ingår i: Epigenetics. - : Taylor & Francis Group. - 1559-2294 .- 1559-2308. ; 17:9, s. 1040-1055
  • Tidskriftsartikel (refereegranskat)abstract
    • Epigenetics may play a central, yet unexplored, role in the profound changes that the maternal immune system undergoes during pregnancy and could be involved in the pregnancy-induced modulation of several autoimmune diseases. We investigated changes in the methylome in isolated circulating CD4(+) T-cells in non-pregnant and pregnant women, during the 1(st) and 2(nd) trimester, using the Illumina Infinium Human Methylation 450K array, and explored how these changes were related to autoimmune diseases that are known to be affected during pregnancy. Pregnancy was associated with several hundreds of methylation differences, particularly during the 2(nd) trimester. A network-based modular approach identified several genes, e.g., CD28, FYN, VAV1 and pathways related to T-cell signalling and activation, highlighting T-cell regulation as a central component of the observed methylation alterations. The identified pregnancy module was significantly enriched for disease-associated methylation changes related to multiple sclerosis, rheumatoid arthritis and systemic lupus erythematosus. A negative correlation between pregnancy-associated methylation changes and disease-associated changes was found for multiple sclerosis and rheumatoid arthritis, diseases that are known to improve during pregnancy whereas a positive correlation was found for systemic lupus erythematosus, a disease that instead worsens during pregnancy. Thus, the directionality of the observed changes is in line with the previously observed effect of pregnancy on disease activity. Our systems medicine approach supports the importance of the methylome in immune regulation of T-cells during pregnancy. Our findings highlight the relevance of using pregnancy as a model for understanding and identifying disease-related mechanisms involved in the modulation of autoimmune diseases.
  •  
3.
  • Badam, Tejaswi V. S., et al. (författare)
  • A validated generally applicable approach using the systematic assessment of disease modules by GWAS reveals a multi-omic module strongly associated with risk factors in multiple sclerosis
  • 2021
  • Ingår i: BMC Genomics. - : BioMed Central. - 1471-2164. ; 22:1
  • Tidskriftsartikel (refereegranskat)abstract
    • 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. 
  •  
4.
  • Badam, Tejaswi Venkata Satya, 1989- (författare)
  • Omic Network Modules in Complex diseases
  • 2021
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • Biological systems encompass various molecular entities such as genes, proteins, and other biological molecules, including interactions among those components. Understanding a given phenotype, the functioning of a cell or tissue, aetiology of disease, or cellular organization, requires accurate measurements of the abundance profiles of these molecular entities in the form of biomedical data. The analysis of the interplay between these different entities at various levels represented in the form of biological network provides a mechanistic understanding of the observed phenotype. In order to study this interplay, there is a requirement of a conceptual and intuitive framework which can model multiple omics such as genome, transcriptome, or a proteome. This can be addressed by application of network-based strategies.Translational bioinformatics deals with the development of analytic and interpretive methods to optimize the transformation of different omics and clinical data to understanding of complex diseases and improving human health. Complex diseases such as multiple sclerosis (MS), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and non-small cell lung cancer (NSCLC) etc., are hypothesized to be a result of a disturbance in the omic networks rendering the healthy cells to be in a state of malfunction. Even though there are numerous methods to layout the relation of the interactions among omics in complex diseases, the output network modules were not clearly interpreted.In this PhD thesis, we showed how different omic data such as transcriptome and methylome can be mapped to the network of interactions to extract highly interconnected gene sets relevant to the disease, so called disease modules. First, we selected common module identification methods and assembled them into a unified framework of the methods implemented in an Rpackage MODifieR (Paper I). Secondly, we showed that the concept of the network modules can be applied on the whole genome sequencing data for developing a tested model for predicting myelosuppressive toxicity (Paper II).Furthermore, we demonstrated that network modules extracted using the methylome data helped identifying several genes that were associated with pregnancy-induced pathways and were enriched for disease-associated methylation changes that were also shared by three auto-immune and inflammatory diseases, namely MS, RA, and SLE (Paper III). Remarkably, those methylation changes correlated with the expected outcome from clinical experience in those diseases. Last, we benchmarked the omic network modules on 19 different complex diseases using both transcriptomic and methylomic data. This led to the identification of a multi-omic MS module that was highly enriched disease-associated genes identified by genome-wide association studies, but also genes associated with the most common environmental risk factors of MS (Paper IV).The application of the network modules concept on different omics is the centrepiece of the research presented in this PhD thesis. The thesis represents the application of omic network modules in complex diseases and how these modules should be integrated and interpreted. In particular, it aimed to show the importance of networks owing to the incomplete knowledge of the genes dysregulated in complex diseases and the contribution of this thesis that provides tools and benchmarks for the methods as well as insights into how a network module can be extracted and interpreted from the omic data in complex diseases.
  •  
5.
  • Bensberg, Maike, et al. (författare)
  • TET2 as a tumor suppressor and therapeutic target in T-cell acute lymphoblastic leukemia
  • 2021
  • Ingår i: Proceedings of the National Academy of Sciences of the United States of America. - : National Academy of Sciences. - 0027-8424 .- 1091-6490. ; 118:34
  • Tidskriftsartikel (refereegranskat)abstract
    • Pediatric T-cell acute lymphoblastic leukemia (T-ALL) is an aggres-sive malignancy resulting from overproduction of immature T-cells in the thymus and is typified by widespread alterations in DNA methyl-ation. As survival rates for relapsed T-ALL remain dismal (10 to 25%), development of targeted therapies to prevent relapse is key to improv-ing prognosis. Whereas mutations in the DNA demethylating enzyme TET2 are frequent in adult T-cell malignancies, TET2 mutations in T-ALL are rare. Here, we analyzed RNA-sequencing data of 321 primary T-ALLs, 20 T-ALL cell lines, and 25 normal human tissues, revealing that TET2 is transcriptionally repressed or silenced in 71% and 17% of T-ALL, respec-tively. Furthermore, we show that TET2 silencing is often associated with hypermethylation of the TET2 promoter in primary T-ALL. Impor-tantly, treatment with the DNA demethylating agent, 5-azacytidine (5-aza), was significantly more toxic to TET2-silenced T-ALL cells and resulted in stable re-expression of the TET2 gene. Additionally, 5-aza led to up-regulation of methylated genes and human endogenous ret-roviruses (HERVs), which was further enhanced by the addition of phys-iological levels of vitamin C, a potent enhancer of TET activity. Together, our results clearly identify 5-aza as a potential targeted therapy for TET2-silenced T-ALL.
  •  
6.
  • Björn, Niclas, 1990-, et al. (författare)
  • Whole-genome sequencing and gene network modules predict gemcitabine/carboplatin-induced myelosuppression in non-small cell lung cancer patients
  • 2020
  • Ingår i: npj Systems Biology and Applications. - : Nature Publishing Group. - 2056-7189. ; 6:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Gemcitabine/carboplatin chemotherapy commonly induces myelosuppression, including neutropenia, leukopenia, and thrombocytopenia. Predicting patients at risk of these adverse drug reactions (ADRs) and adjusting treatments accordingly is a long-term goal of personalized medicine. This study used whole-genome sequencing (WGS) of blood samples from 96 gemcitabine/carboplatin-treated non-small cell lung cancer (NSCLC) patients and gene network modules for predicting myelosuppression. Association of genetic variants in PLINK found 4594, 5019, and 5066 autosomal SNVs/INDELs with p ≤ 1 × 10−3 for neutropenia, leukopenia, and thrombocytopenia, respectively. Based on the SNVs/INDELs we identified the toxicity module, consisting of 215 unique overlapping genes inferred from MCODE-generated gene network modules of 350, 345, and 313 genes, respectively. These module genes showed enrichment for differentially expressed genes in rat bone marrow, human bone marrow, and human cell lines exposed to carboplatin and gemcitabine (p < 0.05). Then using 80% of the patients as training data, random LASSO reduced the number of SNVs/INDELs in the toxicity module into a feasible prediction model consisting of 62 SNVs/INDELs that accurately predict both the training and the test (remaining 20%) data with high (CTCAE 3–4) and low (CTCAE 0–1) maximal myelosuppressive toxicity completely, with the receiver-operating characteristic (ROC) area under the curve (AUC) of 100%. The present study shows how WGS, gene network modules, and random LASSO can be used to develop a feasible and tested model for predicting myelosuppressive toxicity. Although the proposed model predicts myelosuppression in this study, further evaluation in other studies is required to determine its reproducibility, usability, and clinical effect.
  •  
7.
  • Björnsson, Bergthor, et al. (författare)
  • Digital twins to personalize medicine
  • 2020
  • Ingår i: Genome Medicine. - : Springer Science and Business Media LLC. - 1756-994X. ; 12:1
  • Forskningsöversikt (refereegranskat)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.
  •  
8.
  • de Weerd, Hendrik A., et al. (författare)
  • MODalyseR—a novel software for inference of disease module hub regulators identified a putative multiple sclerosis regulator supported by independent eQTL data
  • 2022
  • Ingår i: Bioinformatics Advances. - : Oxford University Press. - 2635-0041. ; 2:1
  • Tidskriftsartikel (refereegranskat)abstract
    • MotivationNetwork-based disease modules have proven to be a powerful concept for extracting knowledge about disease mechanisms, predicting for example disease risk factors and side effects of treatments. Plenty of tools exist for the purpose of module inference, but less effort has been put on simultaneously utilizing knowledge about regulatory mechanisms for predicting disease module hub regulators.ResultsWe developed MODalyseR, a novel software for identifying disease module regulators and reducing modules to the most disease-associated genes. This pipeline integrates and extends previously published software packages MODifieR and ComHub and hereby provides a user-friendly network medicine framework combining the concepts of disease modules and hub regulators for precise disease gene identification from transcriptomics data. To demonstrate the usability of the tool, we designed a case study for multiple sclerosis that revealed IKZF1 as a promising hub regulator, which was supported by independent ChIP-seq data.Availability and implementationMODalyseR is available as a Docker image at https://hub.docker.com/r/ddeweerd/modalyser with user guide and installation instructions found at https://gustafsson-lab.gitlab.io/MODalyseR/.Supplementary informationSupplementary data are available at Bioinformatics Advances online.
  •  
9.
  • de Weerd, Hendrik A., et al. (författare)
  • MODifieR : an ensemble R package for inference of disease modules from transcriptomics networks
  • 2020
  • Ingår i: Bioinformatics. - : Oxford University Press. - 1367-4803 .- 1367-4811 .- 1460-2059. ; 36:12, s. 3918-3919
  • Tidskriftsartikel (refereegranskat)abstract
    • MOTIVATION: Complex diseases are due to the dense interactions of many disease-associated factors that dysregulate genes that in turn form 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: 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.
  •  
10.
  • de Weerd, Hendrik Arnold, 1986- (författare)
  • Novel methods and software for disease module inference
  • 2023
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • Cellular organization is believed to be modular, meaning cellular functions are carried out by modules composed of clusters of genes, proteins and metabolites that are interconnected, co-regulated or physically interacting. In turn, these modules interact together and thereby form complex networks that taken together is considered to be the interactome. Modern high-throughput biological techniques have made high-scale accurate quantification of these biological molecules possible, the so called omics. The simultaneous measurement of these molecules enables a picture of the state of a cell at a resolution that was never before possible. Mapping these measurements aids greatly to elucidate a network structure of interactions. The ever growing size of public repositories for omics data has ushered in the advent of biology as a (big) data science and opens the door for data hungry machine learning approaches in biology. Complex diseases are multi-factorial and arise from a combination of genetic, environmental and lifestyle factors. Additionally, diagnosis and treatment is complicated by the fact that these genetic, environmental and lifestyle factors can vary between patients and may or may not give rise to different disease phenotypes that still classify as the same disease. Genetically, there is substantial heterogeneity among patients and therefore the emergence of a disease phenotype cannot be attributed to a single genetic mutation but rather to a combination of various mutations that may vary from patient to patient. As complex diseases can have different root causes but give rise to a similar disease phenotype, the implication is that different root causes perturb similar components in the interactome. Most of the work in this thesis is aimed at developing methods and computational pipelines to identify, analyze and evaluate these perturbed disease specific sub-networks in the interactome, so called disease modules. We started by collecting popular disease module inference methods and combined them in a unified framework, an R package called MODifieR (Paper I). The package uses standardized inputs and outputs, allowing for a more user-friendly way of running multiple disease module inference methods and the combining of modules. Next, we benchmarked the MODifieR methods on a compendium of transcriptomic and methylomic datasets and combined transcriptomic and methylomic disease modules for Multiple Sclerosis (MS) to a highly disease-relevant module greatly enriched with known risk factors for MS (Paper II). Subsequently, we extended the functionality of MODifieR with software for transcription factor hub detection in gene regulatory networks in a new framework with a graphical user interface, MODalyseR. We used MODalyseR to find upstream regulators and identified IKZF1 as an important upstream regulator for MS (Paper III). Lastly, we used the growing large-scale repositories of gene expression data to train a Variational Auto Encoder (VAE) to compress and decompress gene expression profiles with the aim of extracting disease modules from the latent space. Utilizing the continues nature of the latent space in VAE’s, we derived the differences in latent space representations between a compendium of complex disease gene expression profiles and matched healthy controls. We then derived disease modules from the decompressed latent space representation of this difference and found the modules highly enriched with disease-associated genes, generally outperforming the gold standard of transcriptomic analysis of diseases, top differentially expressed genes (Paper IV). To conclude, the main scientific contribution of this thesis lies in the development of software and methods for improving disease module inference, the evaluation of existing inference methods, the creation of new analysis workflows for multi-omics modules, and the introduction of a deep learning-based approach to the disease module inference toolkit. 
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-10 av 39
Typ av publikation
tidskriftsartikel (28)
doktorsavhandling (8)
forskningsöversikt (2)
annan publikation (1)
Typ av innehåll
refereegranskat (29)
övrigt vetenskapligt/konstnärligt (9)
Författare/redaktör
Gustafsson, Mika (22)
Ernerudh, Jan (8)
Jenmalm, Maria (8)
Gustafsson, Mika, 19 ... (7)
Hellberg, Sandra (6)
Lubovac-Pilav, Zelmi ... (6)
visa fler...
Benson, Mikael (5)
Olsson, Tomas (5)
Nestor, Colm (5)
Martinez, David (5)
Rundquist, Olof (5)
Kockum, Ingrid (4)
Gustafsson, Mika, As ... (4)
Åkesson, Julia (4)
Altafini, Claudio (3)
Raffetseder, Johanna (3)
Khademi, Mohsen (3)
Ernerudh, Jan, 1952- (3)
de Weerd, Hendrik A. (3)
Zhang, Huan (3)
Gawel, Danuta (3)
Li, Xinxiu (3)
Lilja, Sandra (3)
Schäfer, Samuel (3)
Gustafsson, Mika, Pr ... (3)
Jörnsten, Rebecka, 1 ... (2)
Lind, Lars (2)
Piehl, Fredrik (2)
Franks, Paul W. (2)
Jenmalm, Maria, 1971 ... (2)
Matussek, Andreas (2)
Tjärnberg, Andreas (2)
Gustafsson, Stefan (2)
Altafini, Claudio, 1 ... (2)
Wilson, James F. (2)
Hayward, Caroline (2)
Polasek, Ozren (2)
Holmes, Michael V. (2)
Bhai Mehta, Ratnesh (2)
Blomberg, Marie (2)
Badam, Tejaswi V. S. (2)
Jagodic, Maja (2)
Badam, Tejaswi Venka ... (2)
Lentini, Antonio (2)
Yang, Jian (2)
Morris, Andrew P. (2)
Vrethem, Magnus (2)
Jung Lee, Eun Jung (2)
Sysoev, Oleg (2)
Svenvik, Maria (2)
visa färre...
Lärosäte
Linköpings universitet (36)
Karolinska Institutet (15)
Högskolan i Skövde (9)
Göteborgs universitet (4)
Kungliga Tekniska Högskolan (3)
Uppsala universitet (3)
visa fler...
Lunds universitet (3)
Örebro universitet (2)
Chalmers tekniska högskola (2)
visa färre...
Språk
Engelska (39)
Forskningsämne (UKÄ/SCB)
Naturvetenskap (27)
Medicin och hälsovetenskap (19)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy