Network


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

Hotspot


Dive into the research topics where Samuel Collombet is active.

Publication


Featured researches published by Samuel Collombet.


Stem cell reports | 2015

C/EBPα Activates Pre-existing and De Novo Macrophage Enhancers during Induced Pre-B Cell Transdifferentiation and Myelopoiesis

Chris van Oevelen; Samuel Collombet; Guillermo P. Vicent; Maarten Hoogenkamp; Cyrille Lepoivre; Aimee I. Badeaux; Lars H. Bussmann; Jose Luis Sardina; Denis Thieffry; Miguel Beato; Yang Shi; Constanze Bonifer; Thomas Graf

Summary Transcription-factor-induced somatic cell conversions are highly relevant for both basic and clinical research yet their mechanism is not fully understood and it is unclear whether they reflect normal differentiation processes. Here we show that during pre-B-cell-to-macrophage transdifferentiation, C/EBPα binds to two types of myeloid enhancers in B cells: pre-existing enhancers that are bound by PU.1, providing a platform for incoming C/EBPα; and de novo enhancers that are targeted by C/EBPα, acting as a pioneer factor for subsequent binding by PU.1. The order of factor binding dictates the upregulation kinetics of nearby genes. Pre-existing enhancers are broadly active throughout the hematopoietic lineage tree, including B cells. In contrast, de novo enhancers are silent in most cell types except in myeloid cells where they become activated by C/EBP factors. Our data suggest that C/EBPα recapitulates physiological developmental processes by short-circuiting two macrophage enhancer pathways in pre-B cells.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Logical modeling of lymphoid and myeloid cell specification and transdifferentiation

Samuel Collombet; Chris van Oevelen; Jose Luis Sardina Ortega; Wassim Abou-Jaoudé; Bruno Di Stefano; Morgane Thomas-Chollier; Thomas Graf; Denis Thieffry

Blood cells are derived from a common set of hematopoietic stem cells, which differentiate into more specific progenitors of the myeloid and lymphoid lineages, ultimately leading to differentiated cells. This developmental process is controlled by a complex regulatory network involving cytokines and their receptors, transcription factors, and chromatin remodelers. Using public data and data from our own molecular genetic experiments (quantitative PCR, Western blot, EMSA) or genome-wide assays (RNA-sequencing, ChIP-sequencing), we have assembled a comprehensive regulatory network encompassing the main transcription factors and signaling components involved in myeloid and lymphoid development. Focusing on B-cell and macrophage development, we defined a qualitative dynamical model recapitulating cytokine-induced differentiation of common progenitors, the effect of various reported gene knockdowns, and the reprogramming of pre-B cells into macrophages induced by the ectopic expression of specific transcription factors. The resulting network model can be used as a template for the integration of new hematopoietic differentiation and transdifferentiation data to foster our understanding of lymphoid/myeloid cell-fate decisions.


PLOS Genetics | 2017

Krox20 hindbrain regulation incorporates multiple modes of cooperation between cis-acting elements

Elodie Thierion; Johan Le Men; Samuel Collombet; Céline Hernandez; Fanny Coulpier; Patrick Torbey; Morgane Thomas-Chollier; Daan Noordermeer; Patrick Charnay; Pascale Gilardi-Hebenstreit

Developmental genes can harbour multiple transcriptional enhancers that act simultaneously or in succession to achieve robust and precise spatiotemporal expression. However, the mechanisms underlying cooperation between cis-acting elements are poorly documented, notably in vertebrates. The mouse gene Krox20 encodes a transcription factor required for the specification of two segments (rhombomeres) of the developing hindbrain. In rhombomere 3, Krox20 is subject to direct positive feedback governed by an autoregulatory enhancer, element A. In contrast, a second enhancer, element C, distant by 70 kb, is active from the initiation of transcription independent of the presence of the KROX20 protein. Here, using both enhancer knock-outs and investigations of chromatin organisation, we show that element C possesses a dual activity: besides its classical enhancer function, it is also permanently required in cis to potentiate the autoregulatory activity of element A, by increasing its chromatin accessibility. This work uncovers a novel, asymmetrical, long-range mode of cooperation between cis-acting elements that might be essential to avoid promiscuous activation of positive autoregulatory elements.


Cell Stem Cell | 2017

Constitutively Active SMAD2/3 Are Broad-Scope Potentiators of Transcription-Factor-Mediated Cellular Reprogramming

Tyson Ruetz; Ulrich Pfisterer; Bruno Di Stefano; James Ashmore; Meryam Beniazza; Tian V. Tian; Daniel F. Kaemena; Luca Tosti; Wenfang Tan; Jonathan R. Manning; Eleni Chantzoura; Daniella Rylander Ottosson; Samuel Collombet; Anna Johnsson; Erez Cohen; Kosuke Yusa; Sten Linnarsson; Thomas Graf; Malin Parmar; Keisuke Kaji

Summary Reprogramming of cellular identity using exogenous expression of transcription factors (TFs) is a powerful and exciting tool for tissue engineering, disease modeling, and regenerative medicine. However, generation of desired cell types using this approach is often plagued by inefficiency, slow conversion, and an inability to produce mature functional cells. Here, we show that expression of constitutively active SMAD2/3 significantly improves the efficiency of induced pluripotent stem cell (iPSC) generation by the Yamanaka factors. Mechanistically, SMAD3 interacts with reprogramming factors and co-activators and co-occupies OCT4 target loci during reprogramming. Unexpectedly, active SMAD2/3 also markedly enhances three other TF-mediated direct reprogramming conversions, from B cells to macrophages, myoblasts to adipocytes, and human fibroblasts to neurons, highlighting broad and general roles for SMAD2/3 as cell-reprogramming potentiators. Our results suggest that co-expression of active SMAD2/3 could enhance multiple types of TF-based cell identity conversion and therefore be a powerful tool for cellular engineering.


Nucleic Acids Research | 2018

RSAT 2018: regulatory sequence analysis tools 20th anniversary

Nga Thi Thuy Nguyen; Bruno Contreras-Moreira; Jaime A Castro-Mondragon; Walter Santana-Garcia; Raul Ossio; Carla Daniela Robles-Espinoza; Mathieu Bahin; Samuel Collombet; Pierre Vincens; Denis Thieffry; Jacques van Helden; Alejandra Medina-Rivera; Morgane Thomas-Chollier

Abstract RSAT (Regulatory Sequence Analysis Tools) is a suite of modular tools for the detection and the analysis of cis-regulatory elements in genome sequences. Its main applications are (i) motif discovery, including from genome-wide datasets like ChIP-seq/ATAC-seq, (ii) motif scanning, (iii) motif analysis (quality assessment, comparisons and clustering), (iv) analysis of regulatory variations, (v) comparative genomics. Six public servers jointly support 10 000 genomes from all kingdoms. Six novel or refactored programs have been added since the 2015 NAR Web Software Issue, including updated programs to analyse regulatory variants (retrieve-variation-seq, variation-scan, convert-variations), along with tools to extract sequences from a list of coordinates (retrieve-seq-bed), to select motifs from motif collections (retrieve-matrix), and to extract orthologs based on Ensembl Compara (get-orthologs-compara). Three use cases illustrate the integration of new and refactored tools to the suite. This Anniversary update gives a 20-year perspective on the software suite. RSAT is well-documented and available through Web sites, SOAP/WSDL (Simple Object Access Protocol/Web Services Description Language) web services, virtual machines and stand-alone programs at http://www.rsat.eu/.


Scientific Data | 2014

Time-resolved gene expression profiling during reprogramming of C/EBPα-pulsed B cells into iPS cells

Bruno Di Stefano; Samuel Collombet; Thomas Graf

The reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) is lengthy and inefficient. The development of a reprogramming system that allows rapid and synchronous reprogramming to pluripotency is imperative for understanding the mechanism of iPSC formation and for future therapeutic applications. We have recently reported that a short expression in mouse primary B cells of the transcription factor C/EBPα before the induction of pluripotency factors increases the iPSC reprogramming efficiency >100-fold, involving 95% of the cells within a week. Here we present a dataset containing the time course of gene expression during this process as determined by microarray and RNA-seq techniques.


PLOS Genetics | 2018

Cooperation, cis-interactions, versatility and evolutionary plasticity of multiple cis-acting elements underlie krox20 hindbrain regulation

Patrick Torbey; Elodie Thierion; Samuel Collombet; Anne de Cian; Carole Desmarquet-Trin-Dinh; Mathilde Dura; Jean-Paul Concordet; Patrick Charnay; Pascale Gilardi-Hebenstreit

Cis-regulation plays an essential role in the control of gene expression, and is particularly complex and poorly understood for developmental genes, which are subject to multiple levels of modulation. In this study, we performed a global analysis of the cis-acting elements involved in the control of the zebrafish developmental gene krox20. krox20 encodes a transcription factor required for hindbrain segmentation and patterning, a morphogenetic process highly conserved during vertebrate evolution. Chromatin accessibility analysis reveals a cis-regulatory landscape that includes 6 elements participating in the control of initiation and autoregulatory aspects of krox20 hindbrain expression. Combining transgenic reporter analyses and CRISPR/Cas9-mediated mutagenesis, we assign precise functions to each of these 6 elements and provide a comprehensive view of krox20 cis-regulation. Three important features emerged. First, cooperation between multiple cis-elements plays a major role in the regulation. Cooperation can surprisingly combine synergy and redundancy, and is not restricted to transcriptional enhancer activity (for example, 4 distinct elements cooperate through different modes to maintain autoregulation). Second, several elements are unexpectedly versatile, which allows them to be involved in different aspects of control of gene expression. Third, comparative analysis of the elements and their activities in several vertebrate species reveals that this versatility is underlain by major plasticity across evolution, despite the high conservation of the gene expression pattern. These characteristics are likely to be of broad significance for developmental genes.


Molecular and Cellular Biology | 2017

A Transcription Factor Pulse Can Prime Chromatin for Heritable Transcriptional Memory

Aimee Iberg-Badeaux; Samuel Collombet; Benoit Laurent; Chris van Oevelen; Kuo-Kai Chin; Denis Thieffry; Thomas Graf; Yang Shi

ABSTRACT Short-term and long-term transcriptional memory is the phenomenon whereby the kinetics or magnitude of gene induction is enhanced following a prior induction period. Short-term memory persists within one cell generation or in postmitotic cells, while long-term memory can survive multiple rounds of cell division. We have developed a tissue culture model to study the epigenetic basis for long-term transcriptional memory (LTTM) and subsequently used this model to better understand the epigenetic mechanisms that enable heritable memory of temporary stimuli. We find that a pulse of transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) induces LTTM on a subset of target genes that survives nine cell divisions. The chromatin landscape at genes that acquire LTTM is more repressed than at those genes that do not exhibit memory, akin to a latent state. We show through chromatin immunoprecipitation (ChIP) and chemical inhibitor studies that RNA polymerase II (Pol II) elongation is important for establishing memory in this model but that Pol II itself is not retained as part of the memory mechanism. More generally, our work reveals that a transcription factor involved in lineage specification can induce LTTM and that failure to rerepress chromatin is one epigenetic mechanism underlying transcriptional memory.


Nature Cell Biology | 2016

C/EBP[alpha] creates elite cells for iPSC reprogramming by upregulating Klf4 and increasing the levels of Lsd1 and Brd4

Bruno Di Stefano; Samuel Collombet; Janus S. Jakobsen; Michael Wierer; Jose Luis Sardina; Andreas Lackner; Ralph Stadhouders; Carolina Segura-Morales; Mirko Francesconi; Francesco Limone; Matthias Mann; Bo T. Porse; Denis Thieffry; Thomas Graf


Nature Genetics | 2018

Transcription factors orchestrate dynamic interplay between genome topology and gene regulation during cell reprogramming

Ralph Stadhouders; Enrique Vidal; François Serra; Bruno Di Stefano; Francois Le Dily; Javier Quilez; Antonio Gomez; Samuel Collombet; Clara Berenguer; Yasmina Cuartero; Jochen Hecht; Guillaume J. Filion; Miguel Beato; Marc A. Marti-Renom; Thomas Graf

Collaboration


Dive into the Samuel Collombet's collaboration.

Top Co-Authors

Avatar

Thomas Graf

Pompeu Fabra University

View shared research outputs
Top Co-Authors

Avatar

Denis Thieffry

École Normale Supérieure

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ralph Stadhouders

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Elodie Thierion

École Normale Supérieure

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick Charnay

École Normale Supérieure

View shared research outputs
Researchain Logo
Decentralizing Knowledge