Kim Berentsen
Radboud University Nijmegen
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Kim Berentsen.
Science | 2014
Sadia Saeed; Jessica Quintin; Hindrik Hd Kerstens; Nagesha A. Rao; Ali Aghajanirefah; Filomena Matarese; Shih-Chin Cheng; Jacqueline M. Ratter; Kim Berentsen; Martijn van der Ent; Nilofar Sharifi; Eva M. Janssen-Megens; Menno Ter Huurne; Amit Mandoli; Tom van Schaik; Aylwin Ng; Frances Burden; Kate Downes; Mattia Frontini; Vinod Kumar; Evangelos J. Giamarellos-Bourboulis; Willem H. Ouwehand; Jos W. M. van der Meer; Leo A. B. Joosten; Cisca Wijmenga; Joost H.A. Martens; Ramnik J. Xavier; Colin Logie; Mihai G. Netea; Hendrik G. Stunnenberg
Introduction Monocytes circulate in the bloodstream for up to 3 to 5 days. Concomitantly, immunological imprinting of either tolerance (immunosuppression) or trained immunity (innate immune memory) determines the functional fate of monocytes and monocyte-derived macrophages, as observed after infection or vaccination. The epigenome, DNase I accessibility, and transcriptome were characterized in purified human circulating monocytes, in vitro differentiated naïve, tolerized (immunosuppression), and trained macrophages (innate immune memory). This allowed the identification of pathways functionally implicated in innate immune memory. This epigenetic signature of human monocyte-to-macrophage differentiation and monocyte training generates hypotheses to understand and manipulate medically relevant immune conditions. Methods Purified circulating monocytes from healthy volunteers were differentiated under the homeostatic macrophage colony-stimulating factor concentrations present in human serum. During the first 24 hours, trained immunity was induced by β-glucan (BG) priming, and postsepsis immunoparalysis was mimicked by exposure to lipopolysaccharide (LPS), generating endotoxin-induced tolerance. Epigenomic profiling of the histone marks H3K4me1, H3K4me3, and H3K27ac, DNase I accessibility, and RNA sequencing were performed at both the start of the experiment (ex vivo monocytes) and at the end of the 6 days of in vitro culture (macrophages). Results Compared with monocytes (Mo), naïve macrophages (Mf ) display a remodeled metabolic enzyme repertoire and attenuated innate inflammatory pathways, most likely necessary to generate functional tissue macrophages. Epigenetic profiling uncovered about 8000 dynamic regions associated with about 11,000 DNase I hypersensitive sites. Changes in histone acetylation identified most dynamic events. Furthermore, these regions of differential histone marks displayed some degree of DNase I accessibility that was already present in monocytes. H3K4me1 mark increased in parallel with de novo H3K27ac deposition at distal regulatory regions; H3K4me1 mark remained even after the loss of H3K27ac, marking decommissioned regulatory elements. β-glucan priming specifically induced about 3000 distal regulatory elements, whereas LPS tolerization induced H3K27ac at about 500 distal regulatory regions. At the transcriptional level, we identified coregulated gene modules during monocyte-to-macrophage differentiation, as well as discordant modules between trained and tolerized cells. These indicate that training likely involves an increased expression of modules expressed in naïve macrophages, including genes that code for metabolic enzymes. On the other hand, endotoxin tolerance involves gene modules that are more active in monocytes than in naïve macrophages. About 12% of known human transcription factors display variation in expression during macrophage differentiation, training, and tolerance. We also observed transcription factor motifs in DNase I hypersensitive sites at condition-specific dynamic epigenomic regions, implying that specific transcription factors are required for trained and tolerized macrophage epigenetic and transcriptional programs. Finally, our analyses and functional validation indicate that the inhibition of cyclic adenosine monophosphate generation blocked trained immunity in vitro and during an in vivo model of lethal Candida albicans infection, abolishing the protective effects of trained immunity. Discussion We documented the importance of epigenetic regulation of the immunological pathways underlying monocyte-to-macrophage differentiation and trained immunity. These dynamic epigenetic elements may inform on potential pharmacological targets that modulate innate immunity. Altogether, we uncovered the epigenetic and transcriptional programs of monocyte differentiation to macrophages that distinguish tolerant and trained macrophage phenotypes, providing a resource to further understand and manipulate immune-mediated responses. A BLUEPRINT of immune cell development To determine the epigenetic mechanisms that direct blood cells to develop into the many components of our immune system, the BLUEPRINT consortium examined the regulation of DNA and RNA transcription to dissect the molecular traits that govern blood cell differentiation. By inducing immune responses, Saeed et al. document the epigenetic changes in the genome that underlie immune cell differentiation. Cheng et al. demonstrate that trained monocytes are highly dependent on the breakdown of sugars in the presence of oxygen, which allows cells to produce the energy needed to mount an immune response. Chen et al. examine RNA transcripts and find that specific cell lineages use RNA transcripts of different length and composition (isoforms) to form proteins. Together, the studies reveal how epigenetic effects can drive the development of blood cells involved in the immune system. Science, this issue 10.1126/science.1251086, 10.1126/science.1250684, 10.1126/science.1251033 Genome-wide approaches analyze human monocyte differentiation in vitro into functional macrophages. Monocyte differentiation into macrophages represents a cornerstone process for host defense. Concomitantly, immunological imprinting of either tolerance or trained immunity determines the functional fate of macrophages and susceptibility to secondary infections. We characterized the transcriptomes and epigenomes in four primary cell types: monocytes and in vitro–differentiated naïve, tolerized, and trained macrophages. Inflammatory and metabolic pathways were modulated in macrophages, including decreased inflammasome activation, and we identified pathways functionally implicated in trained immunity. β-glucan training elicits an exclusive epigenetic signature, revealing a complex network of enhancers and promoters. Analysis of transcription factor motifs in deoxyribonuclease I hypersensitive sites at cell-type–specific epigenetic loci unveiled differentiation and treatment-specific repertoires. Altogether, we provide a resource to understand the epigenetic changes that underlie innate immunity in humans.
Cell | 2016
William Astle; Heather Elding; Tao Jiang; Dave Allen; Dace Ruklisa; Alice L. Mann; Daniel Mead; Heleen Bouman; Fernando Riveros-Mckay; Myrto Kostadima; John J. Lambourne; Suthesh Sivapalaratnam; Kate Downes; Kousik Kundu; Lorenzo Bomba; Kim Berentsen; John R. Bradley; Louise C. Daugherty; Olivier Delaneau; Kathleen Freson; Stephen F. Garner; Luigi Grassi; Jose A. Guerrero; Matthias Haimel; Eva M. Janssen-Megens; Anita M. Kaan; Mihir Anant Kamat; Bowon Kim; Amit Mandoli; Jonathan Marchini
Summary Many common variants have been associated with hematological traits, but identification of causal genes and pathways has proven challenging. We performed a genome-wide association analysis in the UK Biobank and INTERVAL studies, testing 29.5 million genetic variants for association with 36 red cell, white cell, and platelet properties in 173,480 European-ancestry participants. This effort yielded hundreds of low frequency (<5%) and rare (<1%) variants with a strong impact on blood cell phenotypes. Our data highlight general properties of the allelic architecture of complex traits, including the proportion of the heritable component of each blood trait explained by the polygenic signal across different genome regulatory domains. Finally, through Mendelian randomization, we provide evidence of shared genetic pathways linking blood cell indices with complex pathologies, including autoimmune diseases, schizophrenia, and coronary heart disease and evidence suggesting previously reported population associations between blood cell indices and cardiovascular disease may be non-causal.
Cell | 2016
Lu Chen; Bing Ge; Francesco Paolo Casale; Louella Vasquez; Tony Kwan; Diego Garrido-Martín; Stephen Watt; Ying Yan; Kousik Kundu; Simone Ecker; Avik Datta; David C. Richardson; Frances Burden; Daniel Mead; Alice L. Mann; José María Fernández; Sophia Rowlston; Steven P. Wilder; Samantha Farrow; Xiaojian Shao; John J. Lambourne; Adriana Redensek; Cornelis A. Albers; Vyacheslav Amstislavskiy; Sofie Ashford; Kim Berentsen; Lorenzo Bomba; Guillaume Bourque; David Bujold; Stephan Busche
Summary Characterizing the multifaceted contribution of genetic and epigenetic factors to disease phenotypes is a major challenge in human genetics and medicine. We carried out high-resolution genetic, epigenetic, and transcriptomic profiling in three major human immune cell types (CD14+ monocytes, CD16+ neutrophils, and naive CD4+ T cells) from up to 197 individuals. We assess, quantitatively, the relative contribution of cis-genetic and epigenetic factors to transcription and evaluate their impact as potential sources of confounding in epigenome-wide association studies. Further, we characterize highly coordinated genetic effects on gene expression, methylation, and histone variation through quantitative trait locus (QTL) mapping and allele-specific (AS) analyses. Finally, we demonstrate colocalization of molecular trait QTLs at 345 unique immune disease loci. This expansive, high-resolution atlas of multi-omics changes yields insights into cell-type-specific correlation between diverse genomic inputs, more generalizable correlations between these inputs, and defines molecular events that may underpin complex disease risk.
Nature Communications | 2016
Marcel Smid; Germán Fg Rodríguez-González; Anieta M. Sieuwerts; Roberto Salgado; Wendy Wj Prager-Van der Smissen; Michelle van der Vlugt-Daane; Anne van Galen; Serena Nik-Zainal; Johan Staaf; Arie B. Brinkman; Marc J. van de Vijver; Andrea L. Richardson; Aquila Fatima; Kim Berentsen; Adam A Butler; Sancha Martin; Helen Davies; Reno Debets; Marion E Meijer-Van Me Gelder; Carolien Deurzen; Gaëtan MacGrogan; Gert Van den Eynden; Colin A. Purdie; Alastair M. Thompson; Carlos Caldas; Paul N. Span; Peter T. Simpson; Sunil R. Lakhani; Steven Van Laere; Christine Desmedt
A recent comprehensive whole genome analysis of a large breast cancer cohort was used to link known and novel drivers and substitution signatures to the transcriptome of 266 cases. Here, we validate that subtype-specific aberrations show concordant expression changes for, for example, TP53, PIK3CA, PTEN, CCND1 and CDH1. We find that CCND3 expression levels do not correlate with amplification, while increased GATA3 expression in mutant GATA3 cancers suggests GATA3 is an oncogene. In luminal cases the total number of substitutions, irrespective of type, associates with cell cycle gene expression and adverse outcome, whereas the number of mutations of signatures 3 and 13 associates with immune-response specific gene expression, increased numbers of tumour-infiltrating lymphocytes and better outcome. Thus, while earlier reports imply that the sheer number of somatic aberrations could trigger an immune-response, our data suggests that substitutions of a particular type are more effective in doing so than others.
Cell Reports | 2016
Amit Mandoli; Abhishek A. Singh; Koen H.M. Prange; Esther Tijchon; Marjolein Oerlemans; René A. M. Dirks; Menno Ter Huurne; Albertus T. J. Wierenga; Eva M. Janssen-Megens; Kim Berentsen; Nilofar Sharifi; Bowon Kim; Filomena Matarese; Luan N. Nguyen; Nina C. Hubner; Nagesha A. Rao; Emile van den Akker; Lucia Altucci; Edo Vellenga; Hendrik G. Stunnenberg; Joost H.A. Martens
The t(8;21) acute myeloid leukemia (AML)-associated oncoprotein AML1-ETO disrupts normal hematopoietic differentiation. Here, we have investigated its effects on the transcriptome and epigenome in t(8,21) patient cells. AML1-ETO binding was found at promoter regions of active genes with high levels of histone acetylation but also at distal elements characterized by low acetylation levels and binding of the hematopoietic transcription factors LYL1 and LMO2. In contrast, ERG, FLI1, TAL1, and RUNX1 bind at all AML1-ETO-occupied regulatory regions, including those of the AML1-ETO gene itself, suggesting their involvement in regulating AML1-ETO expression levels. While expression of AML1-ETO in myeloid differentiated induced pluripotent stem cells (iPSCs) induces leukemic characteristics, overexpression increases cell death. We find that expression of wild-type transcription factors RUNX1 and ERG in AML is required to prevent this oncogene overexpression. Together our results show that the interplay of the epigenome and transcription factors prevents apoptosis in t(8;21) AML cells.
Cancer Research | 2016
J.W.M. Martens; M. Smid; G. Rodriguez-Gonzalez; A.M. Sieuwerts; W.J.C. Prager-Van der Smissen; M.V. Van der Vlugt-Daane; A van Galen; Serena Nik-Zainal; Johan Staaf; Arie B. Brinkman; M.J. van de Vijver; Andrea L. Richardson; Kim Berentsen; Carlos Caldas; Adam Butler; Sancha Martin; Hd Davies; Reno Debets; M.E. Meijer-van Gelder; C. H. M. van Deurzen; M.R. Ramakrishna; Markus Ringnér; Alain Viari; Ewan Birney; Anne Lise Børresen-Dale; Henk Stunnenberg; Michael R. Stratton; John A. Foekens
Background: Bladder cancer affects >70,000 patients annually in the United States. Despite its high incidence, therapeutic options are limited in early or late stage. We wanted to identify key metabolic pathways that were altered in bladder cancer development and progression. Experimental Design: We performed global metabolomics profiling of benign urothelium, high-grade non-muscle invasive bladder cancer and advanced, muscle-invasive bladder cancer using GC-MS and LC-MS platforms. This analysis was coupled with publicly available data on transcriptomics of key enzymes, to determine pathways that may be suitable for future therapeutics development. Results: Categorical pathways globally dysregulated in cancer relative to benign urothelium included glucose, TCA cycle, lipid, amino acid and nucleotide pathways. Bladder cancers demonstrated Warburg metabolism, with elevated glucose utilization to drive glycolysis and sorbitol pathway intermediates. Elevated late TCA cycle intermediates, coupled with higher levels of amino acids and dipeptides, suggest the possibility of anaplerotic activity in bladder cancer as a mechanism to sustain energy production. Medium and long chain fatty acids were produced at the expense of dicarboxylic fatty acids. Muscle-invasive bladder cancers showed enhanced use of COX and LOX metabolomics pathways and a possible role for inflammation in regulating NAD+ synthesis in muscle-invasive bladder cancer. Transcriptomic profiling validated that the majority of metabolomics pathway alterations corresponded to gene expression changes of enzymes responsible for metabolite production. Conclusions: This study identifies multiple parallel metabolomics changes unique to non-muscle invasive and muscle-invasive bladder cancer that can be used to justify testing novel therapeutics targeting metabolic pathways in bladder cancer. Citation Format: Divya Sahu, Yair Lotan, Bryan Wittmann, Bruce Neri, Donna Hansel. Metabolomics analysis reveals distinct profiles of non-muscle invasive and muscle-invasive bladder cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2.
bioRxiv | 2017
Daniel Rico; Joost H.A. Martens; Kate Downes; Enrique Carrillo-de-Santa-Pau; Vera Pancaldi; Alessandra Breschi; David C. Richardson; Simon Heath; Sadia Saeed; Mattia Frontini; Lu Chen; Stephen Watt; Fabian Müller; Laura Clarke; Hindrik Hd Kerstens; Steven P. Wilder; Emilio Palumbo; Sarah Djebali; Emanuele Rainieri; Angelika Merkel; Anna Esteve-Codina; Marc Sultan; Alena van Bommel; Marta Gut; Marie-Laure Yaspo; Miriam Rubio; Jose Maria Fernandez; Anthony Attwood; Victor de la Torre; Romina Royo
Neutrophils and monocytes provide a first line of defense against infections as part of the innate immune system. Here we report the integrated analysis of transcriptomic and epigenetic landscapes for circulating monocytes and neutrophils with the aim to enable downstream interpretation and functional validation of key regulatory elements in health and disease. We collected RNA-seq data, ChIP-seq of six histone modifications and of DNA methylation by bisulfite sequencing at base pair resolution from up to 6 individuals per cell type. Chromatin segmentation analyses suggested that monocytes have a higher number of cell-specific enhancer regions (4-fold) compared to neutrophils. This highly plastic epigenome is likely indicative of the greater differentiation potential of monocytes into macrophages, dendritic cells and osteoclasts. In contrast, most of the neutrophil-specific features tend to be characterized by repressed chromatin, reflective of their status as terminally differentiated cells. Enhancers were the regions where most of differences in DNA methylation between cells were observed, with monocyte-specific enhancers being generally hypomethylated. Monocytes show a substantially higher gene expression levels than neutrophils, in line with epigenomic analysis revealing that gene more active elements in monocytes. Our analyses suggest that the overexpression of c-Myc in monocytes and its binding to monocyte-specific enhancers could be an important contributor to these differences. Altogether, our study provides a comprehensive epigenetic chart of chromatin states in primary human neutrophils and monocytes, thus providing a valuable resource for studying the regulation of the human innate immune system.
bioRxiv | 2018
Luigi Grassi; Farzin Pourfarzad; Sebastian Ullrich; Angelika Merkel; Felipe Were; Enrique Carrillo de Santa Pau; Guoqiang Yi; Ida H. Hiemstra; Anton Tool; Erik Mul; Juliane Perner; Eva M. Janssen-Megens; Kim Berentsen; Hinri Kerstens; Ehsan Habibi; Marta Gut; Marie-Laure Yaspo; Matthias Linser; Ernesto Lowy; Avik Datta; Laura Clarke; Paul Flicek; Martin Vingron; Dirk Roos; Timo K. van den Berg; Simon Heath; Daniel Rico; Mattia Frontini; Myrto Kostadima; Ivo Gut
Neutrophils are short-lived blood cells that play a critical role in host defense against infections. To better comprehend neutrophil functions and their regulation, we provide a complete epigenetic and functional overview of their differentiation stages from bone marrow-residing progenitors to mature circulating cells. Integration of epigenetic and transcriptome dynamics reveals an enforced regulation of differentiation, through cellular functions such as: release of proteases, respiratory burst, cell cycle regulation and apoptosis. We observe an early establishment of the cytotoxic capability, whilst the signaling components that activate antimicrobial mechanisms are transcribed at later stages, outside the bone marrow, thus preventing toxic effects in the bone marrow niche. Altogether, these data reveal how the developmental dynamics of the epigenetic landscape orchestrate the daily production of large number of neutrophils required for innate host defense and provide a comprehensive overview of the epigenomes of differentiating human neutrophils. Key points Dynamic acetylation enforces human neutrophil progenitor differentiation. Neutrophils cytotoxic capability is established early at the (pro)myelocyte stage. Coordinated signaling component expression prevents unwanted toxic effects to the bone marrow niche.
Oncotarget | 2018
Abhishek A. Singh; Francesca Petraglia; Angela Nebbioso; Guoqiang Yi; Mariarosaria Conte; Sergio Valente; Amit Mandoli; Lucia Scisciola; Rik G.H. Lindeboom; Hinri Kerstens; Eva M. Janssen-Megens; Farzin Pourfarzad; Ehsan Habibi; Kim Berentsen; Bowon Kim; Colin Logie; Simon Heath; Albertus T. J. Wierenga; Laura Clarke; Paul Flicek; Joop H. Jansen; Taco W. Kuijpers; Marie-Laure Yaspo; Véronique Della Valle; Olivier Bernard; Ivo Gut; Edo Vellenga; Hendrik G. Stunnenberg; Antonello Mai; Lucia Altucci
Epigenomic alterations have been associated with both pathogenesis and progression of cancer. Here, we analyzed the epigenome of two high-risk APL (hrAPL) patients and compared it to non-high-risk APL cases. Despite the lack of common genetic signatures, we found that human hrAPL blasts from patients with extremely poor prognosis display specific patterns of histone H3 acetylation, specifically hyperacetylation at a common set of enhancer regions. In addition, unique profiles of the repressive marks H3K27me3 and DNA methylation were exposed in high-risk APLs. Epigenetic comparison with low/intermediate-risk APLs and AMLs revealed hrAPL-specific patterns of histone acetylation and DNA methylation, suggesting these could be further developed into markers for clinical identification. The epigenetic drug MC2884, a newly generated general HAT/EZH2 inhibitor, induces apoptosis of high-risk APL blasts and reshapes their epigenomes by targeting both active and repressive marks. Together, our analysis uncovers distinctive epigenome signatures of hrAPL patients, and provides proof of concept for use of epigenome profiling coupled to epigenetic drugs to ‘personalize’ precision medicine.
Cell Reports | 2018
Luigi Grassi; Farzin Pourfarzad; Sebastian Ullrich; Angelika Merkel; Felipe Were; Enrique Carrillo-de-Santa-Pau; Guoqiang Yi; Ida H. Hiemstra; Anton Tool; Erik Mul; Juliane Perner; Eva M. Janssen-Megens; Kim Berentsen; Hinri Kerstens; Ehsan Habibi; Marta Gut; Marie-Laure Yaspo; Matthias Linser; Ernesto Lowy; Avik Datta; Laura Clarke; Paul Flicek; Martin Vingron; Dirk Roos; Timo K. van den Berg; Simon Heath; Daniel Rico; Mattia Frontini; Myrto Kostadima; Ivo Gut
Summary Neutrophils are short-lived blood cells that play a critical role in host defense against infections. To better comprehend neutrophil functions and their regulation, we provide a complete epigenetic overview, assessing important functional features of their differentiation stages from bone marrow-residing progenitors to mature circulating cells. Integration of chromatin modifications, methylation, and transcriptome dynamics reveals an enforced regulation of differentiation, for cellular functions such as release of proteases, respiratory burst, cell cycle regulation, and apoptosis. We observe an early establishment of the cytotoxic capability, while the signaling components that activate these antimicrobial mechanisms are transcribed at later stages, outside the bone marrow, thus preventing toxic effects in the bone marrow niche. Altogether, these data reveal how the developmental dynamics of the chromatin landscape orchestrate the daily production of a large number of neutrophils required for innate host defense and provide a comprehensive overview of differentiating human neutrophils.