Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hana Imrichova is active.

Publication


Featured researches published by Hana Imrichova.


PLOS Computational Biology | 2014

iRegulon: from a gene list to a gene regulatory network using large motif and track collections.

Rekin's Janky; Annelien Verfaillie; Hana Imrichova; Bram Van de Sande; Laura Standaert; Valerie Christiaens; Gert Hulselmans; Koen Herten; Marina Naval Sanchez; Delphine Potier; Dmitry Svetlichnyy; Zeynep Kalender Atak; Mark Fiers; Jean-Christophe Marine; Stein Aerts

Identifying master regulators of biological processes and mapping their downstream gene networks are key challenges in systems biology. We developed a computational method, called iRegulon, to reverse-engineer the transcriptional regulatory network underlying a co-expressed gene set using cis-regulatory sequence analysis. iRegulon implements a genome-wide ranking-and-recovery approach to detect enriched transcription factor motifs and their optimal sets of direct targets. We increase the accuracy of network inference by using very large motif collections of up to ten thousand position weight matrices collected from various species, and linking these to candidate human TFs via a motif2TF procedure. We validate iRegulon on gene sets derived from ENCODE ChIP-seq data with increasing levels of noise, and we compare iRegulon with existing motif discovery methods. Next, we use iRegulon on more challenging types of gene lists, including microRNA target sets, protein-protein interaction networks, and genetic perturbation data. In particular, we over-activate p53 in breast cancer cells, followed by RNA-seq and ChIP-seq, and could identify an extensive up-regulated network controlled directly by p53. Similarly we map a repressive network with no indication of direct p53 regulation but rather an indirect effect via E2F and NFY. Finally, we generalize our computational framework to include regulatory tracks such as ChIP-seq data and show how motif and track discovery can be combined to map functional regulatory interactions among co-expressed genes. iRegulon is available as a Cytoscape plugin from http://iregulon.aertslab.org.


Nature Communications | 2015

Decoding the regulatory landscape of melanoma reveals TEADS as regulators of the invasive cell state

Annelien Verfaillie; Hana Imrichova; Zeynep Kalender Atak; Michael Dewaele; Florian Rambow; Gert Hulselmans; Christiaens; Dmitry Svetlichnyy; Flavie Luciani; Van den Mooter L; Claerhout S; Mark Fiers; Fabrice Journé; Ghanem Elias Ghanem; Carl Herrmann; Georg Halder; Jean-Christophe Marine; Stein Aerts

Transcriptional reprogramming of proliferative melanoma cells into a phenotypically distinct invasive cell subpopulation is a critical event at the origin of metastatic spreading. Here we generate transcriptome, open chromatin and histone modification maps of melanoma cultures; and integrate this data with existing transcriptome and DNA methylation profiles from tumour biopsies to gain insight into the mechanisms underlying this key reprogramming event. This shows thousands of genomic regulatory regions underlying the proliferative and invasive states, identifying SOX10/MITF and AP-1/TEAD as regulators, respectively. Knockdown of TEADs shows a previously unrecognized role in the invasive gene network and establishes a causative link between these transcription factors, cell invasion and sensitivity to MAPK inhibitors. Using regulatory landscapes and in silico analysis, we show that transcriptional reprogramming underlies the distinct cellular states present in melanoma. Furthermore, it reveals an essential role for the TEADs, linking it to clinically relevant mechanisms such as invasion and resistance.


eLife | 2015

Transcription factor MITF and remodeller BRG1 define chromatin organisation at regulatory elements in melanoma cells

Patrick Laurette; Thomas Strub; Dana Koludrovic; Céline Keime; Stéphanie Le Gras; Hannah Seberg; Eric Van Otterloo; Hana Imrichova; Robert Siddaway; Stein Aerts; Robert A. Cornell; Gabrielle Mengus; Irwin Davidson

Microphthalmia-associated transcription factor (MITF) is the master regulator of the melanocyte lineage. To understand how MITF regulates transcription, we used tandem affinity purification and mass spectrometry to define a comprehensive MITF interactome identifying novel cofactors involved in transcription, DNA replication and repair, and chromatin organisation. We show that MITF interacts with a PBAF chromatin remodelling complex comprising BRG1 and CHD7. BRG1 is essential for melanoma cell proliferation in vitro and for normal melanocyte development in vivo. MITF and SOX10 actively recruit BRG1 to a set of MITF-associated regulatory elements (MAREs) at active enhancers. Combinations of MITF, SOX10, TFAP2A, and YY1 bind between two BRG1-occupied nucleosomes thus defining both a signature of transcription factors essential for the melanocyte lineage and a specific chromatin organisation of the regulatory elements they occupy. BRG1 also regulates the dynamics of MITF genomic occupancy. MITF-BRG1 interplay thus plays an essential role in transcription regulation in melanoma. DOI: http://dx.doi.org/10.7554/eLife.06857.001


Nucleic Acids Research | 2015

i-cisTarget 2015 update: generalized cis-regulatory enrichment analysis in human, mouse and fly

Hana Imrichova; Gert Hulselmans; Zeynep Kalender Atak; Delphine Potier; Stein Aerts

i-cisTarget is a web tool to predict regulators of a set of genomic regions, such as ChIP-seq peaks or co-regulated/similar enhancers. i-cisTarget can also be used to identify upstream regulators and their target enhancers starting from a set of co-expressed genes. Whereas the original version of i-cisTarget was focused on Drosophila data, the 2015 update also provides support for human and mouse data. i-cisTarget detects transcription factor motifs (position weight matrices) and experimental data tracks (e.g. from ENCODE, Roadmap Epigenomics) that are enriched in the input set of regions. As experimental data tracks we include transcription factor ChIP-seq data, histone modification ChIP-seq data and open chromatin data. The underlying processing method is based on a ranking-and-recovery procedure, allowing accurate determination of enrichment across heterogeneous datasets, while also discriminating direct from indirect target regions through a ‘leading edge’ analysis. We illustrate i-cisTarget on various Ewing sarcoma datasets to identify EWS-FLI1 targets starting from ChIP-seq, differential ATAC-seq, differential H3K27ac and differential gene expression data. Use of i-cisTarget is free and open to all, and there is no login requirement. Address: http://gbiomed.kuleuven.be/apps/lcb/i-cisTarget.


Nature Methods | 2017

SCENIC: single-cell regulatory network inference and clustering

Sara Aibar; Thomas Moerman; Vân Anh Huynh-Thu; Hana Imrichova; Gert Hulselmans; Florian Rambow; Jean-Christophe Marine; Pierre Geurts; Jan Aerts; Joost van den Oord; Zeynep Kalender Atak; Jasper Wouters; Stein Aerts

We present SCENIC, a computational method for simultaneous gene regulatory network reconstruction and cell-state identification from single-cell RNA-seq data (http://scenic.aertslab.org). On a compendium of single-cell data from tumors and brain, we demonstrate that cis-regulatory analysis can be exploited to guide the identification of transcription factors and cell states. SCENIC provides critical biological insights into the mechanisms driving cellular heterogeneity.


Genome Research | 2016

Multiplex enhancer-reporter assays uncover unsophisticated TP53 enhancer logic

Annelien Verfaillie; Dmitry Svetlichnyy; Hana Imrichova; Kristofer Davie; Mark Fiers; Zeynep Kalender Atak; Gert Hulselmans; Valerie Christiaens; Stein Aerts

Transcription factors regulate their target genes by binding to regulatory regions in the genome. Although the binding preferences of TP53 are known, it remains unclear what distinguishes functional enhancers from nonfunctional binding. In addition, the genome is scattered with recognition sequences that remain unoccupied. Using two complementary techniques of multiplex enhancer-reporter assays, we discovered that functional enhancers could be discriminated from nonfunctional binding events by the occurrence of a single TP53 canonical motif. By combining machine learning with a meta-analysis of TP53 ChIP-seq data sets, we identified a core set of more than 1000 responsive enhancers in the human genome. This TP53 cistrome is invariably used between cell types and experimental conditions, whereas differences among experiments can be attributed to indirect nonfunctional binding events. Our data suggest that TP53 enhancers represent a class of unsophisticated cell-autonomous enhancers containing a single TP53 binding site, distinct from complex developmental enhancers that integrate signals from multiple transcription factors.


PLOS Computational Biology | 2015

Identification of High-Impact cis-Regulatory Mutations Using Transcription Factor Specific Random Forest Models

Dmitry Svetlichnyy; Hana Imrichova; Mark Fiers; Zeynep Kalender Atak; Stein Aerts

Cancer genomes contain vast amounts of somatic mutations, many of which are passenger mutations not involved in oncogenesis. Whereas driver mutations in protein-coding genes can be distinguished from passenger mutations based on their recurrence, non-coding mutations are usually not recurrent at the same position. Therefore, it is still unclear how to identify cis-regulatory driver mutations, particularly when chromatin data from the same patient is not available, thus relying only on sequence and expression information. Here we use machine-learning methods to predict functional regulatory regions using sequence information alone, and compare the predicted activity of the mutated region with the reference sequence. This way we define the Predicted Regulatory Impact of a Mutation in an Enhancer (PRIME). We find that the recently identified driver mutation in the TAL1 enhancer has a high PRIME score, representing a “gain-of-target” for MYB, whereas the highly recurrent TERT promoter mutation has a surprisingly low PRIME score. We trained Random Forest models for 45 cancer-related transcription factors, and used these to score variations in the HeLa genome and somatic mutations across more than five hundred cancer genomes. Each model predicts only a small fraction of non-coding mutations with a potential impact on the function of the encompassing regulatory region. Nevertheless, as these few candidate driver mutations are often linked to gains in chromatin activity and gene expression, they may contribute to the oncogenic program by altering the expression levels of specific oncogenes and tumor suppressor genes.


Molecular & Cellular Proteomics | 2015

Combined Proteomics and Transcriptomics Identifies Carboxypeptidase B1 and Nuclear Factor κB (NF-κB) Associated Proteins as Putative Biomarkers of Metastasis in Low Grade Breast Cancer

Pavel Bouchal; Monika Dvořáková; Theodoros Roumeliotis; Zbyněk Bortlíček; Ivana Ihnatová; Iva Procházková; Jenny Ho; Josef Maryáš; Hana Imrichova; Eva Budinská; Rostislav Vyzula; Spiros D. Garbis; Bořivoj Vojtěšek; Rudolf Nenutil

Current prognostic factors are insufficient for precise risk-discrimination in breast cancer patients with low grade breast tumors, which, in disagreement with theoretical prognosis, occasionally form early lymph node metastasis. To identify markers for this group of patients, we employed iTRAQ-2DLC-MS/MS proteomics to 24 lymph node positive and 24 lymph node negative grade 1 luminal A primary breast tumors. Another group of 48 high-grade tumors (luminal B, triple negative, Her-2 subtypes) was also analyzed to investigate marker specificity for grade 1 luminal A tumors. From the total of 4405 proteins identified (FDR<5%), the top 65 differentially expressed together with 30 previously identified and control markers were analyzed also at transcript level. Increased levels of carboxypeptidase B1 (CPB1), PDZ and LIM domain protein 2 (PDLIM2), and ring finger protein 25 (RNF25) were associated specifically with lymph node positive grade 1 tumors, whereas stathmin 1 (STMN1) and thymosin beta 10 (TMSB10) associated with aggressive tumor phenotype also in high grade tumors at both protein and transcript level. For CPB1, these differences were also observed by immunohistochemical analysis on tissue microarrays. Up-regulation of putative biomarkers in lymph node positive (versus negative) luminal A tumors was validated by gene expression analysis of an independent published data set (n = 343) for CPB1 (p = 0.00155), PDLIM2 (p = 0.02027) and RELA (p = 0.00015). Moreover, statistically significant connections with patient survival were identified in another public data set (n = 1678). Our findings indicate unique pro-metastatic mechanisms in grade 1 tumors that can include up-regulation of CPB1, activation of NF-κB pathway and changes in cell survival and cytoskeleton. These putative biomarkers have potential to identify the specific minor subpopulation of breast cancer patients with low grade tumors who are at higher than expected risk of recurrence and who would benefit from more intensive follow-up and may require more personalized therapy.


Current protocols in human genetics | 2015

iRegulon and i‐cisTarget: Reconstructing Regulatory Networks Using Motif and Track Enrichment

Annelien Verfaillie; Hana Imrichova; Rekin's Janky; Stein Aerts

Gene expression profiling is often used to identify genes that are co‐expressed in a biological process or disease. Downstream analyses of co‐expressed gene sets using bioinformatics methods can reveal candidate transcription factors (TF) that co‐regulate these genes, based on the presence of shared TF binding sites. Drawing gene regulatory networks that connect TFs to their predicted target genes can uncover gene modules that implement a particular function. Here, we describe several protocols to analyze any set of co‐expressed genes using iRegulon and i‐cisTarget. These tools perform regulatory sequence analysis (motif discovery) and integrate and mine large collections of existing regulatory data, such as ChIP‐Seq, DHS‐seq, and FAIRE‐seq (track discovery). While iRegulon focuses on sets of co‐expressed genes, i‐cisTarget also analyses genomic regions as input. The following protocols describe how to install and use these tools, how to interpret the obtained results, and will thus help to create meaningful regulatory networks.


Genome Medicine | 2017

Identification of cis-regulatory mutations generating de novo edges in personalized cancer gene regulatory networks

Zeynep Kalender Atak; Hana Imrichova; Dmitry Svetlichnyy; Gert Hulselmans; Valerie Christiaens; Joke Reumers; Hugo Ceulemans; Stein Aerts

The identification of functional non-coding mutations is a key challenge in the field of genomics. Here we introduce μ-cisTarget to filter, annotate and prioritize cis-regulatory mutations based on their putative effect on the underlying “personal” gene regulatory network. We validated μ-cisTarget by re-analyzing the TAL1 and LMO1 enhancer mutations in T-ALL, and the TERT promoter mutation in melanoma. Next, we re-sequenced the full genomes of ten cancer cell lines and used matched transcriptome data and motif discovery to identify master regulators with de novo binding sites that result in the up-regulation of nearby oncogenic drivers. μ-cisTarget is available from http://mucistarget.aertslab.org.

Collaboration


Dive into the Hana Imrichova's collaboration.

Top Co-Authors

Avatar

Stein Aerts

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Gert Hulselmans

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Zeynep Kalender Atak

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Annelien Verfaillie

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Valerie Christiaens

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Dmitry Svetlichnyy

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Mark Fiers

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Delphine Potier

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge