Network


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

Hotspot


Dive into the research topics where Bence Daniel is active.

Publication


Featured researches published by Bence Daniel.


Genes & Development | 2014

The active enhancer network operated by liganded RXR supports angiogenic activity in macrophages

Bence Daniel; Gergely Nagy; Nasun Hah; Attila Horvath; Zsolt Czimmerer; Szilard Poliska; Tibor Gyuris; Jiri Keirsse; Conny Gysemans; Jo A. Van Ginderachter; Balint L. Balint; Ronald M. Evans; Endre Barta; Laszlo Nagy

RXR signaling is predicted to have a major impact in macrophages, but neither the biological consequence nor the genomic basis of its ligand activation is known. Comprehensive genome-wide studies were carried out to map liganded RXR-mediated transcriptional changes, active binding sites, and cistromic interactions in the context of the macrophage genome architecture. The macrophage RXR cistrome has 5200 genomic binding sites, which are not impacted by ligand. Active enhancers are characterized by PU.1 binding, an increase of enhancer RNA, and P300 recruitment. Using these features, 387 liganded RXR-bound enhancers were linked to 226 genes, which predominantly reside in CTCF/cohesin-limited functional domains. These findings were molecularly validated using chromosome conformation capture (3C) and 3C combined with sequencing (3C-seq), and we show that selected long-range enhancers communicate with promoters via stable or RXR-induced loops and that some of the enhancers interact with each other, forming an interchromosomal network. A set of angiogenic genes, including Vegfa, has liganded RXR-controlled enhancers and provides the macrophage with a novel inducible program.


Immunity | 2016

Macrophage PPARγ, a Lipid Activated Transcription Factor Controls the Growth Factor GDF3 and Skeletal Muscle Regeneration

Tamas Varga; Rémi Mounier; Andreas Patsalos; Péter Gogolák; Matthew Peloquin; Attila Horvath; Attila Pap; Bence Daniel; Gergely Nagy; Éva Pintye; Szilard Poliska; Sylvain Cuvellier; Sabrina Ben Larbi; Brian E. Sansbury; Matthew Spite; Chester W. Brown; Bénédicte Chazaud; Laszlo Nagy

Tissue regeneration requires inflammatory and reparatory activity of macrophages. Macrophages detect and eliminate the damaged tissue and subsequently promote regeneration. This dichotomy requires the switch of effector functions of macrophages coordinated with other cell types inside the injured tissue. The gene regulatory events supporting the sensory and effector functions of macrophages involved in tissue repair are not well understood. Here we show that the lipid activated transcription factor, PPARγ, is required for proper skeletal muscle regeneration, acting in repair macrophages. PPARγ controls the expression of the transforming growth factor-β (TGF-β) family member, GDF3, which in turn regulates the restoration of skeletal muscle integrity by promoting muscle progenitor cell fusion. This work establishes PPARγ as a required metabolic sensor and transcriptional regulator of repair macrophages. Moreover, this work also establishes GDF3 as a secreted extrinsic effector protein acting on myoblasts and serving as an exclusively macrophage-derived regeneration factor in tissue repair.


Molecular and Cellular Biology | 2014

Ligand Binding Shifts Highly Mobile Retinoid X Receptor to the Chromatin-Bound State in a Coactivator-Dependent Manner, as Revealed by Single-Cell Imaging

Peter Brazda; Jan Krieger; Bence Daniel; Dávid Jónás; Tibor Szekeres; Jörg Langowski; Katalin Tóth; Laszlo Nagy; György Vámosi

ABSTRACT Retinoid X receptor (RXR) is a promiscuous nuclear receptor forming heterodimers with several other receptors, which activate different sets of genes. Upon agonist treatment, the occupancy of its genomic binding regions increased, but only a modest change in the number of sites was revealed by chromatin immunoprecipitation followed by sequencing, suggesting a rather static behavior. However, such genome-wide and biochemical approaches do not take into account the dynamic behavior of a transcription factor. Therefore, we characterized the nuclear dynamics of RXR during activation in single cells on the subsecond scale using live-cell imaging. By applying fluorescence recovery after photobleaching and fluorescence correlation spectroscopy (FCS), techniques with different temporal and spatial resolutions, a highly dynamic behavior could be uncovered which is best described by a two-state model (slow and fast) of receptor mobility. In the unliganded state, most RXRs belonged to the fast population, leaving ∼15% for the slow, chromatin-bound fraction. Upon agonist treatment, this ratio increased to ∼43% as a result of an immediate and reversible redistribution. Coactivator binding appears to be indispensable for redistribution and has a major contribution to chromatin association. A nuclear mobility map recorded by light sheet microscopy-FCS shows that the ligand-induced transition from the fast to the slow population occurs throughout the nucleus. Our results support a model in which RXR has a distinct, highly dynamic nuclear behavior and follows hit-and-run kinetics upon activation.


Genome Medicine | 2016

The IL-4/STAT6 signaling axis establishes a conserved microRNA signature in human and mouse macrophages regulating cell survival via miR-342-3p

Zsolt Czimmerer; Tamas Varga; Mate Kiss; Cesaré Ovando Vázquez; Quang Minh Doan-Xuan; Dominik Rückerl; Sudhir Gopal Tattikota; Xin Yan; Zsuzsanna S. Nagy; Bence Daniel; Szilard Poliska; Attila Horvath; Gergely Nagy; Éva Várallyay; Matthew N. Poy; Judith E. Allen; Zsolt Bacsó; Cei Abreu-Goodger; Laszlo Nagy

BackgroundIL-4-driven alternative macrophage activation and proliferation are characteristic features of both antihelminthic immune responses and wound healing in contrast to classical macrophage activation, which primarily occurs during inflammatory responses. The signaling pathways defining the genome-wide microRNA expression profile as well as the cellular functions controlled by microRNAs during alternative macrophage activation are largely unknown. Hence, in the current work we examined the regulation and function of IL-4-regulated microRNAs in human and mouse alternative macrophage activation.MethodsWe utilized microarray-based microRNA profiling to detect the dynamic expression changes during human monocyte–macrophage differentiation and IL-4-mediated alternative macrophage activation. The expression changes and upstream regulatory pathways of selected microRNAs were further investigated in human and mouse in vitro and in vivo models of alternative macrophage activation by integrating small RNA-seq, ChIP-seq, ChIP-quantitative PCR, and gene expression data. MicroRNA-controlled gene networks and corresponding functions were identified using a combination of transcriptomic, bioinformatic, and functional approaches.ResultsThe IL-4-controlled microRNA expression pattern was identified in models of human and mouse alternative macrophage activation. IL-4-dependent induction of miR-342-3p and repression of miR-99b along with miR-125a-5p occurred in both human and murine macrophages in vitro. In addition, a similar expression pattern was observed in peritoneal macrophages of Brugia malayi nematode-implanted mice in vivo. By using IL4Rα- and STAT6-deficient macrophages, we were able to show that IL-4-dependent regulation of miR-342-3p, miR-99b, and miR-125a-5p is mediated by the IL-4Rα–STAT6 signaling pathway. The combination of gene expression studies and chromatin immunoprecipitation experiments demonstrated that both miR-342-3p and its host gene, EVL, are coregulated directly by STAT6. Finally, we found that miR-342-3p is capable of controlling macrophage survival through targeting an anti-apoptotic gene network including Bcl2l1.ConclusionsOur findings identify a conserved IL-4/STAT6-regulated microRNA signature in alternatively activated human and mouse macrophages. Moreover, our study indicates that miR-342-3p likely plays a pro-apoptotic role in such cells, thereby providing a negative feedback arm to IL-4-dependent macrophage proliferation.


Immunobiology | 2013

A novel method to predict regulatory regions based on histone mark landscapes in macrophages

Gergely Nagy; Bence Daniel; Dávid Jónás; Laszlo Nagy; Endre Barta

Macrophages as phagocytes and professional antigen presenting cells play critical roles in both innate and adaptive immunity. Main transcription factors acting during their differentiation and function are known, but the behavior and co-operation of these factors still remained unexplored. We introduce a new approach to map nucleosome-free regions using exclusively active enhancer and core promoter marking histone modification ChIP-seq data. We could detect approximately 56,000 potential active enhancers/promoters showing different lengths and histone modification shapes. Beside the highly enriched PU.1 and C/EBP sites, we could also detect binding sites for RUNX and AP-1, as well as for the MiT (MITF-TFE) family and MEF2 proteins. The PU.1 and C/EBP transcription factors are known for transforming cells into macrophages. The other transcription factors found in this study can play a role in macrophages as well, since it is known that the MiT family proteins are responsible for phagocytic activity and the MEF2 proteins specify monocytic differentiation over the granulocyte direction. Our results imply that this method can provide novel information about transcription factor organization at enhancers and core promoters as well as about the histone modifications surrounding regulatory regions in any immune or other cell types.


BMC Genomics | 2013

Reprogramming of lysosomal gene expression by interleukin-4 and Stat6

Louise M Brignull; Zsolt Czimmerer; Hafida Saidi; Bence Daniel; Izabel Villela; Nathan W. Bartlett; Sebastian L. Johnston; Lisiane B. Meira; Laszlo Nagy; Axel Nohturfft

BackgroundLysosomes play important roles in multiple aspects of physiology, but the problem of how the transcription of lysosomal genes is coordinated remains incompletely understood. The goal of this study was to illuminate the physiological contexts in which lysosomal genes are coordinately regulated and to identify transcription factors involved in this control.ResultsAs transcription factors and their target genes are often co-regulated, we performed meta-analyses of array-based expression data to identify regulators whose mRNA profiles are highly correlated with those of a core set of lysosomal genes. Among the ~50 transcription factors that rank highest by this measure, 65% are involved in differentiation or development, and 22% have been implicated in interferon signaling. The most strongly correlated candidate was Stat6, a factor commonly activated by interleukin-4 (IL-4) or IL-13. Publicly available chromatin immunoprecipitation (ChIP) data from alternatively activated mouse macrophages show that lysosomal genes are overrepresented among Stat6-bound targets. Quantification of RNA from wild-type and Stat6-deficient cells indicates that Stat6 promotes the expression of over 100 lysosomal genes, including hydrolases, subunits of the vacuolar H+ ATPase and trafficking factors. While IL-4 inhibits and activates different sets of lysosomal genes, Stat6 mediates only the activating effects of IL-4, by promoting increased expression and by neutralizing undefined inhibitory signals induced by IL-4.ConclusionsThe current data establish Stat6 as a broadly acting regulator of lysosomal gene expression in mouse macrophages. Other regulators whose expression correlates with lysosomal genes suggest that lysosome function is frequently re-programmed during differentiation, development and interferon signaling.


Nature Medicine | 2018

Transcriptional regulation of macrophage cholesterol efflux and atherogenesis by a long noncoding RNA

Tamer Sallam; Marius Jones; Brandon J. Thomas; Xiaohui Wu; Thomas Gilliland; Kevin Qian; Ascia Eskin; David Casero; Zhengyi Zhang; Jaspreet Sandhu; David Salisbury; Prashant Rajbhandari; Mete Civelek; Cynthia Hong; Ayaka Ito; Xin Liu; Bence Daniel; Aldons J. Lusis; Julian P. Whitelegge; Laszlo Nagy; Antonio Castrillo; Stephen T. Smale; Peter Tontonoz

Nuclear receptors regulate gene expression in response to environmental cues, but the molecular events governing the cell type specificity of nuclear receptors remain poorly understood. Here we outline a role for a long noncoding RNA (lncRNA) in modulating the cell type–specific actions of liver X receptors (LXRs), sterol-activated nuclear receptors that regulate the expression of genes involved in cholesterol homeostasis and that have been causally linked to the pathogenesis of atherosclerosis. We identify the lncRNA MeXis as an amplifier of LXR-dependent transcription of the gene Abca1, which is critical for regulation of cholesterol efflux. Mice lacking the MeXis gene show reduced Abca1 expression in a tissue-selective manner. Furthermore, loss of MeXis in mouse bone marrow cells alters chromosome architecture at the Abca1 locus, impairs cellular responses to cholesterol overload, and accelerates the development of atherosclerosis. Mechanistic studies reveal that MeXis interacts with and guides promoter binding of the transcriptional coactivator DDX17. The identification of MeXis as a lncRNA modulator of LXR-dependent gene expression expands understanding of the mechanisms underlying cell type–selective actions of nuclear receptors in physiology and disease.


Immunity | 2018

The Transcription Factor STAT6 Mediates Direct Repression of Inflammatory Enhancers and Limits Activation of Alternatively Polarized Macrophages

Zsolt Czimmerer; Bence Daniel; Attila Horvath; Dominik Rückerl; Gergely Nagy; Mate Kiss; Matthew Peloquin; Marietta Margit Budai; Ixchelt Cuaranta-Monroy; Zoltan Simandi; László Steiner; Béla Nagy; Szilard Poliska; Csaba Bankó; Zsolt Bacsó; Ira G. Schulman; Sascha Sauer; Jean-François Deleuze; Judith E. Allen; Szilvia Benko; Laszlo Nagy

Summary The molecular basis of signal‐dependent transcriptional activation has been extensively studied in macrophage polarization, but our understanding remains limited regarding the molecular determinants of repression. Here we show that IL‐4‐activated STAT6 transcription factor is required for the direct transcriptional repression of a large number of genes during in vitro and in vivo alternative macrophage polarization. Repression results in decreased lineage‐determining transcription factor, p300, and RNA polymerase II binding followed by reduced enhancer RNA expression, H3K27 acetylation, and chromatin accessibility. The repressor function of STAT6 is HDAC3 dependent on a subset of IL‐4‐repressed genes. In addition, STAT6‐repressed enhancers show extensive overlap with the NF‐&kgr;B p65 cistrome and exhibit decreased responsiveness to lipopolysaccharide after IL‐4 stimulus on a subset of genes. As a consequence, macrophages exhibit diminished inflammasome activation, decreased IL‐1&bgr; production, and pyroptosis. Thus, the IL‐4‐STAT6 signaling pathway establishes an alternative polarization‐specific epigenenomic signature resulting in dampened macrophage responsiveness to inflammatory stimuli. Graphical Abstract Figure. No Caption available. HighlightsIL‐4‐activated STAT6 acts as a transcriptional repressor in macrophagesIL‐4‐STAT6‐repressed enhancers associate with reduced LDTF and p300 bindingInflammatory responsiveness of the IL‐4‐repressed enhancers is attenuatedIL‐4 limits the LPS‐induced inflammasome activation, IL‐1&bgr; production, and pyroptosis &NA; The molecular bases of repressive transcriptional mechanisms contributing to macrophage polarization are not well understood. Czimmerer et al. show that in alternatively polarized macrophages, IL‐4‐activated STAT6 represses a large set of enhancers modulating the transcriptional program. STAT6‐repressed enhancers are characterized by reduced chromatin accessibility, eRNA expression, LDTF, and p300 binding. IL‐4‐STAT6‐mediated repression limits the inflammatory responsiveness including inflammasome activation, IL‐1&bgr; production, and pyroptosis. Thus, the IL4‐STAT6 pathway establishes an epigenomic signature to selectively repress the macrophage inflammation program.


FEBS Letters | 2014

The intriguing complexities of mammalian gene regulation: How to link enhancers to regulated genes. Are we there yet?

Bence Daniel; Gergely Nagy; Laszlo Nagy

The information encoded in genomes supports the differentiation and function of the more than 200 unique cell types, which exist in various mammalian species. The major mechanism driving cellular differentiation and specification is differential gene expression regulation. Cis‐acting enhancers and silencers appear to have key roles in regulating the expression of mammalian genes. However, these cis‐acting elements are often located very far away from the regulated gene. Therefore, it is hard to find all of them and link them to the regulated gene. An intriguing and unresolved issue of the field is to identify all of the enhancers of a particular gene and link these short regulatory sequences to the genes they regulate and thus, reliably identify gene regulatory enhancer networks. Recent advances in molecular biological methods coupled with Next‐Generation Sequencing (NGS) technologies have opened up new possibilities in this area of genomics. In this review we summarize the technological advances, bioinformatics challenges and the potential molecular mechanisms allowing the construction of enhancer networks operating in specific cell types and/or activated by various signals.


Biochimica et Biophysica Acta | 2016

Transcriptional control of transglutaminase 2 expression in mouse apoptotic thymocytes

Katalin Sándor; Bence Daniel; Bea Kiss; Fruzsina Kovács; Zsuzsa Szondy

Transglutaminase 2 (TGM2) is a ubiquitously expressed multifunctional protein, which participates in various biological processes including thymocyte apoptosis. As a result, the transcriptional regulation of the gene is complex and must depend on the cell type. Previous studies from our laboratory have shown that in dying thymocytes the expression of Tgm2 is induced by external signals derived from engulfing macrophages, such as retinoids, transforming growth factor (TGF)-β and adenosine, the latter triggering the adenylate cyclase signaling pathway. The existence of TGF-β and retinoid responsive elements in the promoter region of Tgm2 has already been reported, but the intergenic regulatory elements participating in the regulation of Tgm2 have not yet been identified. Here we used publicly available results from DNase I hypersensitivity analysis followed by deep sequencing and chromatin immunoprecipitation followed by deep sequencing against CCCTC-binding factor (CTCF), H3K4me3, H3K4me1 and H3K27ac to map a putative regulatory element set for Tgm2 in thymocytes. By measuring eRNA expressions of these putative enhancers in retinoid, rTGF-β or dibutiryl cAMP-exposed thymocytes we determined which of them are functional. By applying ChIP-qPCR against SMAD4, retinoic acid receptor, retinoid X receptor, cAMP response element binding protein, P300 and H3K27ac under the same conditions, we identified two enhancers of Tgm2, which seem to act as integrators of the TGF-β, retinoid and adenylate cyclase signaling pathways in dying thymocytes. Our study describes a novel strategy to identify and characterize the signal-specific functional enhancer set of a gene by integrating genome-wide datasets and measuring the production of enhancer specific RNA molecules.

Collaboration


Dive into the Bence Daniel's collaboration.

Top Co-Authors

Avatar

Laszlo Nagy

University of Debrecen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mate Kiss

University of Debrecen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sascha Sauer

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge