top of page
Nature2023_Arnould_Rocher_Legube.PNG

In this article in Nature, we report that the production of DSBs drive the ATM-dependent formation of a new chromatin compartment, called compartment D (for DSB induced). This D compartment forms via the clustering of damaged topologically associating domains, decorated with γH2AX and 53BP1, and by a mechanism that is consistent with polymer–polymer phase separation rather than liquid–liquid phase separation. The D compartment arises mostly in G1 phase, is independent of cohesin and is enhanced after inhibition of DNA-PK or R-loop accumulation. Importantly, R-loop-enriched DNA-damage-responsive genes physically localize to the D compartment, and this contributes to their optimal activation, providing a function for DSB clustering in the DNA damage response. However, DSB-induced chromosome reorganization comes at the expense of an increased rate of translocations, also observed in cancer genomes. Overall, we characterize how DSB-induced compartmentalization orchestrates the DNA damage response and highlight the critical impact of chromosome architecture in genomic instability.

2023
2022
BLM POLD3 SETX and R-loops DSB.JPG

In this Nature Communications article we uncover a function for the Bloom RecQ DNA helicase (BLM) in TC-DSBR in human cells. BLM is recruited in a transcription dependent-manner at DSBs where it fosters resection, RAD51 binding and accurate Homologous Recombination repair. However, in an R-loop dissolution-deficient background, we find that BLM promotes cell death. We report that upon excessive RNA:DNA hybrid accumulation, DNA synthesis is enhanced at DSBs, in a manner that depends on BLM and POLD3. Altogether our work unveils a role for BLM at DSBs in active chromatin, and highlights the toxic potential of RNA:DNA hybrids that accumulate at transcription-associated DSBs.

2021
DSB in active loci.JPG

This review article in DNA repair  discusses about recent work regarding DSBs occurring in transcribed loci. It provides an updated view of the “Transcription-Coupled DSB Repair” (TC-DSBR) pathway(s) that are mounted at DSBs occurring in loci transcribed by RNA Polymerase I (RNAPI) or RNA Polymerase II  (RNAPII), highlighting differences and common features, as  well as  yet  unanswered questions.

JANUS FACED.PNG

This review article in Nature Cell Biology  discusses about the formation and function of  R-loops  that accumulate in cis to DSBs, especially those induced in transcriptionally active loci. Whether R-loops actively participate in DSB repair or are detrimental by-products that must be removed to avoid genome instability is also addressed.

loop extrusion.JPG

This article in Nature shows that topologically associating domains are functional units of the DNA damage response, and are instrumental for the correct establishment of γH2AX–53BP1 chromatin domains in a manner that involves one-sided cohesin-mediated loop extrusion on both sides of the DSB. We propose a model in which H2AX-containing nucleosomes are rapidly phosphorylated as they actively pass by DSB-anchored cohesin. Our work highlights the importance of chromosome conformation in the maintenance of genome integrity and demonstrates the establishment of a chromatin modification by loop extrusion.

XRCC4 RAD51 BLESS.JPG
Analyzing Homologous Recombination at a Genome-Wide Level

This chapter is part of the Methods in Molecular Biology book chapter on Homologous recombination.

It present our method to perform the genome-wide mapping of Rad51 by ChIP-seq in the DIvA cell system, where multiple DSBs can be induced in a controlled manner by the AsiSI restriction enzyme. Importantly this method can also be implemented using either alternative ways to induce annotated DSBs (I-PpoI, CRISPR/Cas9) or using chemical agents that produce DSB in a non-random manner across the genome (such as etoposide).

BLM BIR.JPG
BLM-dependent Break-Induced Replication handles DSBs in transcribed chromatin upon impaired RNA:DNA hybrids dissolution

This manuscript was added on bioRxiv.

It shows a critical function of the Bloom RecQ DNA helicase (BLM) in transcription-coupled DSB repair (TC-DSBR) in human cells. By being recruited in a transcription-dependent manner, BLM fosters resection at damaged active genes, promotes RAD51 binding and is thus required Homologous Recombination repair. Yet in a context where R-loops are not removed, BLM switches from promoting Homologous Recombination to promoting Break-Induced Replication (BIR). This pathway switch strongly impairs cell viability. Altogether we uncover a novel role for BLM in BIR at DSBs in active chromatin, and highlights the toxic potential of RNA:DNA hybrids at transcription-associated DSBs.
 

tools.JPG
Studying DNA Double-Strand Break Repair: An Ever-Growing Toolbox

This review in Frontiers in Molecular Biosciences written in collaboration with Pablo Huertas and Sérgio Fernandes de Almeida discusses our current knowledge of the molecular determinants and mechanistic details of the DDR. This review essentially focuses on the  development of ground-breaking experimental tools which help improving our understanding of the DDR. This review also addresses the pros and cons of each technique that is currently used to induce DSB, and analyse the DDR.

m6A modififed R-loops.JPG
m6A RNA modification as a new player in R-loop regulation

This News and Views in Nature Genetics discusses a new study that shows that N6-methyladenosine (m6A) modification on the RNA moieties regulates the formation and genome integrity of these hybrids. This finding opens a new avenue of research on how RNA modifications (the ‘epitranscriptome’) can help control genome maintenance.

secret life of chromosome.JPG
The Secret Life of Chromosome Loops upon DNA Double-Strand Break

This review in the Journal of Molecular Biology is on the impact of DSB on chromatin composition and chromosome architecture. Our knowledge on the topic is very sparse despite the recent development of various tools to induce DSB at annotated loci, compatible with next-generation sequencing-based approaches. In this review, we discuss the influence of initial and DSB-induced chromatin conformation and the strong potential of 3C-based technologies to decipher the contribution of chromosome architecture during DSB repair.

rDNA DSB repair.jpg
A cohesin/HUSH- and LINC-dependent pathway controls ribosomal DNA double-strand break repair

This article in Genes and Developement explains how a particularly unstable locus of our genome, the ribosomal DNA (rDNA), is repaired. DSBs within the rDNA locus induce both rDNA transcriptional repression and nucleolar segregation, but the link between the two events remains unclear. Here we found that DSBs induced on rDNA trigger transcriptional repression in a cohesin- and HUSH (human silencing hub) complex-dependent manner throughout the cell cycle. In S/G2 cells, transcriptional repression is further followed by extended resection within the interior of the nucleolus, DSB mobilization at the nucleolar periphery within nucleolar caps, and repair by homologous recombination. We showed that nuclear envelope invaginations frequently connect the nucleolus and that rDNA DSB mobilization, but not transcriptional repression, involves the nuclear envelope-associated LINC complex and the actin pathway.

roadblocks or repair intermediate.JPG
Non-canonical DNA/RNA structures during Transcription-Coupled Double-Strand Break Repair: Roadblocks or Bona fide repair intermediates?

This review in DNA repair was written in collaboration with Kyle Miller and discusses about nucleic acid structures in DNA repair. It is now well understood that DNA does not systematically assemble into a canonical double helix but can also accommodate other structures including DNA hairpins, G-quadruplexes and RNA:DNA hybrids. Notably, these non-canonical DNA structures form preferentially at transcriptionally active loci. Acting as replication roadblocks and being targeted by multiple machineries, these structures weaken the genome and render it prone to damage, including DSB. In addition, secondary structures also further accumulate upon DSB formation. Here we discuss the potential functions of pre-existing or de novo formed nucleic acid structures, as bona fide repair intermediates or repair roadblocks, especially during Transcription-Coupled DNA Double-Strand Break repair (TC-DSBR).

chomatin modifications and DSB mibility
A Snapshot on the Cis Chromatin Response to DNA Double-Strand Breaks.

This review in Trends in Genetics talks about the chromatin alteration observed at various scales around DSBs, in a manner that relates to the repair pathway used, hence defining a 'repair histone code'. The recent advances regarding our knowledge of the chromatin landscape induced in cis around DSBs, with an emphasis on histone post-translational modifications and histone variants are being disccused.

Ancre 2018

2018

2019
nanoblade.JPG
Genome editing in primary cells and in vivo using viral-derived
Nanoblades loaded with Cas9-sgRNA ribonucleoproteins

Emiliano Ricci's team engineered murine leukemia virus-like particles loaded with Cas9-sgRNA ribonucleoproteins (Nanoblades) to induce efficient genome-editing in cell lines and primary cells including human induced pluripotent stem cells, human hematopoietic stem cells and mouse bone-marrow cells. Nanoblades preparation process is simple, relatively inexpensive and can be easily implemented in any laboratory equipped for cellular biology.

thomas clouaire histone mapping

In this Molecular Cell paper, we provide the most comprehensive picture of the chromatin landscape set up at DSBs and identify NHEJ- and HR-specific chromatin events. This study revealed the existence of a DSB-induced monoubiquitination-to-acetylation switch on histone H2B lysine 120, likely mediated by the SAGA complex, as well as higher-order signaling at HR-repaired DSBs whereby histone H1 is evicted while ubiquitin and 53BP1 accumulate over the entire γH2AX domains.

SETX-Rloop.JPG

In this Nature communication paper, we report that senataxin is recruited at DSB when they occur in transcriptionally active loci. Genome-wide mapping unveiled that RNA:DNA hybrids accumulate on DSB-flanking chromatin but display a narrow, DSB-induced, depletion near DNA ends coinciding with senataxin binding. Although neither required for resection nor for timely repair of DSBs, senataxin was found to promote Rad51 recruitment, to minimize illegitimate rejoining of distant DNA ends and to sustain cell viability following DSB production in active genes. Our data suggest that senataxin functions at DSBs in order to limit translocations and ensure cell viability, providing new insights on AOA2/ALS4 neuropathies

DYNLL1.JPG

Dipanjan Chowdhury's team identified DYNLL1 as an inhibitor of DNA end resection. The loss of DYNLL1 enables DNA end resection and restores homologous recombination in BRCA1-mutant cells, thereby inducing resistance to platinum drugs and inhibitors of poly(ADP-ribose) polymerase. In cells, DYNLL1 limits nucleolytic degradation of DNA ends by associating with the DNA end-resection machinery. In vitro, DYNLL1 binds directly to MRE11 to limit its end-resection activity. Therefore, they infer that DYNLL1 is an important anti-resection factor that influences genomic stability and responses to DNA-damaging chemotherapy.

DSB predictions.JPG

Raphaël Mourad devised a computational approach to predict DSBs using the epigenomic and chromatin context, for which public data are readily available from the ENCODE project. They achieved excellent prediction accuracy at high resolution. They identify chromatin accessibility, activity, and long-range contacts as the best predictors

2017

Ancre 2017
DSB go for broke.JPG

This review article in Nucleus discusses of the mechanisms that could sustain clustering. Translocations are dramatic genomic rearrangements due to aberrant rejoining of distant DNA ends that can trigger cancer onset and progression. Translocations frequently occur in genes, yet the mechanisms underlying their formation remain poorly understood. One potential mechanism involves DNA Double Strand Break mobility and juxtaposition (i.e. clustering), an event that has been intensively debated over the past decade. Using Capture Hi-C, we recently found that DSBs do in fact cluster in human nuclei but only when induced in transcriptionally active genes. Notably, we found that clustering of damaged genes is regulated by cell cycle progression and coincides with damage persistency. 

ATM is on duty.JPG

This review in DNA repair is on the ATM protein which has been known for decades as the main kinase mediating the DNA Double-Strand Break Response (DDR). Extensive studies have revealed its dual role in locally promoting detection and repair of DSBs as well as in activating global DNA damage checkpoints. However, recent studies pinpoint additional unanticipated functions for ATM in modifying both the local chromatin landscape and the global chromosome organization, more particularly at persistent breaks. Given the emergence of a novel and unexpected class of DSBs prevalently arising in transcriptionally active genes and intrinsically difficult to repair, a specific role of ATM at refractory DSBs could be an important and so far overlooked feature of Ataxia Telangiectasia (A-T) a severe disorder associated with ATM mutations.

active gene need special care.JPG

This review in the Journal of Molecular Biology is about the current understanding of the crosstalk between transcription and double-strand break repair, from the reasons underlying the intrinsic fragility of genes to the mechanisms that restore the integrity of damaged transcription units.

DSB clustering.JPG

In this NSMB paper, we used a high-throughput chromosome conformation capture assay (capture Hi-C) to investigate clustering of DSBs induced at defined loci in the human genome. The results unambiguously demonstrated that DSBs cluster, but only when they are induced within transcriptionally active genes. Clustering of damaged genes occurs primarily during the G1 cell-cycle phase and coincides with delayed repair. Moreover, DSB clustering depends on the MRN complex as well as the Formin 2 (FMN2) nuclear actin organizer and the linker of nuclear and cytoplasmic skeleton (LINC) complex, thus suggesting that active mechanisms promote clustering. This work reveals that, when damaged, active genes, compared with the rest of the genome, exhibit a distinctive behavior, remaining largely unrepaired and clustered in G1, and being repaired via homologous recombination in postreplicative cells.

DSB hit the road.JPG

This review in current Opinion in Cell Biology focuses on DSB movement. In the past decade, large-scale movements of DNA double strand breaks (DSBs) have repeatedly been identified following DNA damage. These mobility events include clustering, anchoring or peripheral movement at subnuclear structures. Recent work suggests roles for motion in homology search and in break sequestration to preclude deleterious outcomes. Yet, the precise functions of these movements still remain relatively obscure, and the same holds true for the determinants. Here we review recent advances in this exciting area of research, and highlight that a recurrent characteristic of mobile DSBs may lie in their inability to undergo rapid repair. A major future challenge remains to understand how DSB mobility impacts on genome integrity.

Kyle miller ZMYND8.JPG

Kyle Miller's team reports the identification of the histone demethylase KDM5A as a key regulator of the bromodomain protein ZMYND8 and NuRD (nucleosome remodeling and histone deacetylation) complex in the DNA damage response. They observe KDM5A-dependent H3K4me3 demethylation within chromatin near DNA double-strand break (DSB) sites. Mechanistically, demethylation of H3K4me3 is required for ZMYND8-NuRD binding to chromatin and recruitment to DNA damage. Functionally, KDM5A deficiency results in impaired transcriptional silencing and repair of DSBs by homologous recombination. Thus, this study identifies a crucial function for KDM5A in demethylating H3K4 to allow ZMYND8-NuRD to operate within damaged chromatin to repair DSBs.

Ancre 2016
TAD ATM chromatin.JPG

This review in Molecular & Cellular Oncology describes the roles of Ataxia telangiectasia mutated (ATM) at the sites of breaks and its ability to modify both the local chromatin landscape and the global chromosome organization in order to promote repair accuracy.

TIP60.JPG

Jacques Côté's team focusses on the NuA4/TIP60 acetyltransferase complex which is a key regulator of genome expression and stability. Here they identified MBTD1 as a stable subunit of the complex, and we reveal that, via a histone reader domain for H4K20me1/2, MBTD1 allows TIP60 to associate with specific gene promoters and to promote the repair of DNA double-strand breaks by homologous recombination. It was previously suggested that TIP60-dependent acetylation of H4 regulates binding of the non-homologous end joining factor 53BP1, which engages chromatin through simultaneous binding of H4K20me2 and H2AK15ub. They found that the TIP60 complex regulates association of 53BP1 partly by competing for H4K20me2 and by regulating H2AK15ub. Ubiquitylation of H2AK15 by RNF168 inhibits chromatin acetylation by TIP60, while this residue can be acetylated by TIP60 in vivo, blocking its ubiquitylation. Altogether, these results uncover an intricate mechanism orchestrated by the TIP60 complex to regulate 53BP1-dependent repair through competitive bivalent binding and modification of chromatin.

Ancre 2015
ATM and DNAPK.JPG

In this article in Cell Reports, we used the DIvA system (DSB inducible via AsiSI) combined with high-resolution mapping and advanced microscopy to uncovered that both ATM and DNA-PKcs spread in cis on a confined region surrounding DSBs, independently of the pathway used for repair. However, once recruited, these kinases exhibit non-overlapping functions on end joining and γH2AX domain establishment. More specifically, we found that ATM is required to ensure the association of multiple DSBs within "repair foci." Our results suggest that ATM acts not only on chromatin marks but also on higher-order chromatin organization to ensure repair accuracy and survival.

topo I induced breaks Sordet and Legube.

Olivier Sordet's team used camptothecin (CPT)-treated serum-starved quiescent cells to induce transcription-blocking Top1cc and show that those DSBs are generated during Top1cc repair from Top1 peptide-linked DNA single-strand breaks generated after Top1 proteolysis and before excision by TDP1. Following DSB induction, ATM activates DNA-PK whose inhibition suppresses H2AX and H2A ubiquitination and the later assembly of activated ATM into nuclear foci. Inhibition of DNA-PK also reduces Top1 ubiquitination and proteolysis as well as resumption of RNA synthesis suggesting that DSB signaling further enhances Top1cc repair. Finally, they show that co-transcriptional DSBs kill quiescent cells. Together, these new findings reveal that DSB production and signaling by transcription-blocking Top1 lesions impact on non-replicating cell fate and provide insights on the molecular pathogenesis of neurodegenerative diseases such as SCAN1 and AT syndromes, which are caused by TDP1 and ATM deficiency, respectively.

ZMYND8 Kyle Miller and Legube.JPG

Kyle Miller's team reports that human bromodomain (BRD)-containing proteins, the primary "readers" of acetylated chromatin, are vital for the DNA damage response (DDR). We discovered that more than one-third of all human BRD proteins change localization in response to DNA damage. We identified ZMYND8 (zinc finger and MYND [myeloid, Nervy, and DEAF-1] domain containing 8) as a novel DDR factor that recruits the nucleosome remodeling and histone deacetylation (NuRD) complex to damaged chromatin. Our data define a transcription-associated DDR pathway mediated by ZMYND8 and the NuRD complex that targets DNA damage, including when it occurs within transcriptionally active chromatin, to repress transcription and promote repair by homologous recombination. Thus, our data identify human BRD proteins as key chromatin modulators of the DDR and provide novel insights into how DNA damage within actively transcribed regions requires chromatin-binding proteins to orchestrate the appropriate response in concordance with the damage-associated chromatin context.

Ancre 2014
aymard 2014.JPG

In this Nature Structural and Molecular Biology paper, we show that transcriptionally active chromatin is preferentially repaired by HR. Using chromatin immunoprecipitation-sequencing (ChIP-seq) to analyze repair of multiple DSBs induced throughout the human genome, we identify an HR-prone subset of DSBs that recruit the HR protein RAD51, undergo resection and rely on RAD51 for efficient repair. These DSBs are located in actively transcribed genes and are targeted to HR repair via the transcription elongation-associated mark trimethylated histone H3 K36. Concordantly, depletion of SETD2, the main H3 K36 trimethyltransferase, severely impedes HR at such DSBs. Our study thereby demonstrates a primary role in DSB repair of the chromatin context in which a break occurs.

chailleux Nature protocols 2014.JPG

In Collaboration with Didier Trouche's team we describe a protocol for quantifying endonuclease-induced DSBs; this quantification is essential to an interpretation of how DSBs are managed and repaired. A biotinylated double-stranded oligonucleotide is ligated to enzyme-cleaved genomic DNA, allowing the purification of the cleaved DNA on streptavidin beads. The extent of cleavage is then quantified either by quantitative PCR (qPCR) at a given site or at multiple sites by genome-wide techniques (e.g., microarrays or high-throughput sequencing). This technique, named ligation-mediated purification, can be performed in 2 d. It is more accurate and sensitive than existing alternative methods, and it is compatible with genome-wide analysis. It allows the amount of endonuclease-mediated breaks to be precisely compared between two conditions or across the genome, thereby giving insight into the influence of a given factor or of various chromatin contexts on local repair parameters.

Tanya Paull 2014.JPG

Tanyal Paull's team developped a novel method to quantitatively measure single-stranded DNA intermediates in human cells and find that the 5' strand at endonuclease-generated break sites is resected up to 3.5 kb in a cell cycle-dependent manner. Depletion of CtIP, Mre11, Exo1 or SOSS1 blocks resection, while depletion of 53BP1, Ku or DNA-dependent protein kinase catalytic subunit leads to increased resection as measured by this method. While 53BP1 negatively regulates DNA end processing, depletion of Brca1 does not, suggesting that the role of Brca1 in HR is primarily to promote Rad51 filament formation, not to regulate end resection.

Jim haber 2014.JPG

Jim Haber's team describe Mec1- and Tel1-dependent phosphorylation of histone H2B at T129 in yeast. γ-H2B formation is impaired by γ-H2AX and its binding partner Rad9. High-density microarray analyses show similar γ-H2AX and γ-H2B distributions, but γ-H2B is absent near telomeres. Both γ-H2AX and γ-H2B are strongly diminished over highly transcribed regions. When transcription of GAL7, GAL10 and GAL1 genes is turned off, γ-H2AX is restored within 5 min, in a Mec1-dependent manner; after reinduction of these genes, γ-H2AX is rapidly lost. Moreover, when a DSB is induced near CEN2, γ-H2AX spreads to all other pericentromeric regions, again depending on Mec1. Our data provide new insights in the function and establishment of phosphorylation events occurring on chromatin after DSB induction

Tim Humphrey 2014.JPG

Tim Humphrey's team describe a role for H3K36 trimethylation in homologous recombination (HR) repair in human cells is defined. They found that depleting SETD2 generates a mutation signature resembling RAD51 depletion at I-SceI-induced DNA double-strand break (DSB) sites, with significantly increased deletions arising through microhomology-mediated end-joining. They established a presynaptic role for SETD2 methyltransferase in HR, where it facilitates the recruitment of C-terminal binding protein interacting protein (CtIP) and promotes DSB resection, allowing Replication Protein A (RPA) and RAD51 binding to DNA damage sites. Furthermore, reducing H3K36me3 levels by overexpressing KDM4A/JMJD2A, an oncogene and H3K36me3/2 demethylase, or an H3.3K36M transgene also reduces HR repair events. They propose that error-free HR repair within H3K36me3-decorated transcriptionally active genomic regions promotes cell homeostasis. Moreover, these findings provide insights as to why oncogenic mutations cluster within the H3K36me3 axis

bottom of page