Network


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

Hotspot


Dive into the research topics where Hannes Lans is active.

Publication


Featured researches published by Hannes Lans.


Journal of Cell Biology | 2009

Heterochromatin protein 1 is recruited to various types of DNA damage

Martijn S. Luijsterburg; Christoffel Dinant; Hannes Lans; Jan Stap; Elzbieta Wiernasz; Saskia Lagerwerf; Daniël O. Warmerdam; Michael Lindh; Maartje C. Brink; Jurek Dobrucki; Jacob A. Aten; Maria Fousteri; Gert Jansen; Nico P. Dantuma; Wim Vermeulen; Leon H.F. Mullenders; Adriaan B. Houtsmuller; Pernette J. Verschure; Roel van Driel

Heterochromatin protein 1 (HP1) family members are chromatin-associated proteins involved in transcription, replication, and chromatin organization. We show that HP1 isoforms HP1-α, HP1-β, and HP1-γ are recruited to ultraviolet (UV)-induced DNA damage and double-strand breaks (DSBs) in human cells. This response to DNA damage requires the chromo shadow domain of HP1 and is independent of H3K9 trimethylation and proteins that detect UV damage and DSBs. Loss of HP1 results in high sensitivity to UV light and ionizing radiation in the nematode Caenorhabditis elegans, indicating that HP1 proteins are essential components of DNA damage response (DDR) systems. Analysis of single and double HP1 mutants in nematodes suggests that HP1 homologues have both unique and overlapping functions in the DDR. Our results show that HP1 proteins are important for DNA repair and may function to reorganize chromatin in response to damage.


Journal of Cell Biology | 2009

Nucleotide excision repair–induced H2A ubiquitination is dependent on MDC1 and RNF8 and reveals a universal DNA damage response

Jurgen A. Marteijn; Simon Bekker-Jensen; Niels Mailand; Hannes Lans; Petra Schwertman; Audrey M. Gourdin; Nico P. Dantuma; Jiri Lukas; Wim Vermeulen

The epigenetic mark indicative of DNA UV damage or double-strand breaks is achieved via a common pathway regardless of the cause of damage.


Epigenetics & Chromatin | 2012

ATP-dependent chromatin remodeling in the DNA-damage response

Hannes Lans; Jurgen A. Marteijn; Wim Vermeulen

The integrity of DNA is continuously challenged by metabolism-derived and environmental genotoxic agents that cause a variety of DNA lesions, including base alterations and breaks. DNA damage interferes with vital processes such as transcription and replication, and if not repaired properly, can ultimately lead to premature aging and cancer. Multiple DNA pathways signaling for DNA repair and DNA damage collectively safeguard the integrity of DNA. Chromatin plays a pivotal role in regulating DNA-associated processes, and is itself subject to regulation by the DNA-damage response. Chromatin influences access to DNA, and often serves as a docking or signaling site for repair and signaling proteins. Its structure can be adapted by post-translational histone modifications and nucleosome remodeling, catalyzed by the activity of ATP-dependent chromatin-remodeling complexes. In recent years, accumulating evidence has suggested that ATP-dependent chromatin-remodeling complexes play important, although poorly characterized, roles in facilitating the effectiveness of the DNA-damage response. In this review, we summarize the current knowledge on the involvement of ATP-dependent chromatin remodeling in three major DNA repair pathways: nucleotide excision repair, homologous recombination, and non-homologous end-joining. This shows that a surprisingly large number of different remodeling complexes display pleiotropic functions during different stages of the DNA-damage response. Moreover, several complexes seem to have multiple functions, and are implicated in various mechanistically distinct repair pathways.


PLOS Genetics | 2010

Involvement of global genome repair, transcription coupled repair, and chromatin remodeling in UV DNA damage response changes during development.

Hannes Lans; Jurgen A. Marteijn; Björn Schumacher; Jan H.J. Hoeijmakers; Gert Jansen; Wim Vermeulen

Nucleotide Excision Repair (NER), which removes a variety of helix-distorting lesions from DNA, is initiated by two distinct DNA damage-sensing mechanisms. Transcription Coupled Repair (TCR) removes damage from the active strand of transcribed genes and depends on the SWI/SNF family protein CSB. Global Genome Repair (GGR) removes damage present elsewhere in the genome and depends on damage recognition by the XPC/RAD23/Centrin2 complex. Currently, it is not well understood to what extent both pathways contribute to genome maintenance and cell survival in a developing organism exposed to UV light. Here, we show that eukaryotic NER, initiated by two distinct subpathways, is well conserved in the nematode Caenorhabditis elegans. In C. elegans, involvement of TCR and GGR in the UV-induced DNA damage response changes during development. In germ cells and early embryos, we find that GGR is the major pathway contributing to normal development and survival after UV irradiation, whereas in later developmental stages TCR is predominantly engaged. Furthermore, we identify four ISWI/Cohesin and four SWI/SNF family chromatin remodeling factors that are implicated in the UV damage response in a developmental stage dependent manner. These in vivo studies strongly suggest that involvement of different repair pathways and chromatin remodeling proteins in UV-induced DNA repair depends on developmental stage of cells.


Molecular Cell | 2013

Enhanced Chromatin Dynamics by FACT Promotes Transcriptional Restart after UV-Induced DNA Damage

Christoffel Dinant; Giannis Ampatziadis-Michailidis; Hannes Lans; Maria Tresini; Anna Lagarou; Małgorzata Grosbart; Arjan F. Theil; Wiggert A. van Cappellen; Hiroshi Kimura; Jiri Bartek; Maria Fousteri; Adriaan B. Houtsmuller; Wim Vermeulen; Jurgen A. Marteijn

Chromatin remodeling is tightly linked to all DNA-transacting activities. To study chromatin remodeling during DNA repair, we established quantitative fluorescence imaging methods to measure the exchange of histones in chromatin in living cells. We show that particularly H2A and H2B are evicted and replaced at an accelerated pace at sites of UV-induced DNA damage. This accelerated exchange of H2A/H2B is facilitated by SPT16, one of the two subunits of the histone chaperone FACT (facilitates chromatin transcription) but largely independent of its partner SSRP1. Interestingly, SPT16 is targeted to sites of UV light-induced DNA damage-arrested transcription and is required for efficient restart of RNA synthesis upon damage removal. Together, our data uncover an important role for chromatin dynamics at the crossroads of transcription and the UV-induced DNA damage response.


Neuron | 2004

G Protein-Coupled Receptor Kinase Function Is Essential for Chemosensation in C. elegans

Hana S. Fukuto; Denise M. Ferkey; Alfonso junior Apicella; Hannes Lans; Tahira Sharmeen; Wei Chen; Robert J. Lefkowitz; Gert Jansen; William R. Schafer; Anne C. Hart

G protein-coupled receptors (GPCRs) mediate diverse signaling processes, including olfaction. G protein-coupled receptor kinases (GRKs) are important regulators of G protein signal transduction that specifically phosphorylate activated GPCRs to terminate signaling. Despite previously described roles for GRKs in GPCR signal downregulation, animals lacking C. elegans G protein-coupled receptor kinase-2 (Ce-grk-2) function are not hypersensitive to odorants. Instead, decreased Ce-grk-2 function in adult sensory neurons profoundly disrupts chemosensation, based on both behavioral analysis and Ca(2+) imaging. Although mammalian arrestin proteins cooperate with GRKs in receptor desensitization, loss of C. elegans arrestin-1 (arr-1) does not disrupt chemosensation. Either overexpression of the C. elegans Galpha subunit odr-3 or loss of eat-16, which encodes a regulator of G protein signaling (RGS) protein, restores chemosensation in Ce-grk-2 mutants. These results demonstrate that loss of GRK function can lead to reduced GPCR signal transduction and suggest an important role for RGS proteins in the regulation of chemosensation.


Nature | 2006

Cell biology: Ageing nucleus gets out of shape

Hannes Lans; Jan H.J. Hoeijmakers

In certain premature-ageing syndromes, the architecture of the cell nucleus is abnormal. An animal model shows similar malformations during normal ageing, corroborating the idea that genome instability underlies ageing.


Nature Communications | 2015

SUMO and ubiquitin-dependent XPC exchange drives nucleotide excision repair

Loes van Cuijk; Gijsbert J. van Belle; Yasemin Turkyilmaz; Sara Lund Poulsen; Roel C. Janssens; Arjan F. Theil; Mariangela Sabatella; Hannes Lans; Niels Mailand; Adriaan B. Houtsmuller; Wim Vermeulen; Jurgen A. Marteijn

XPC recognizes UV-induced DNA lesions and initiates their removal by nucleotide excision repair (NER). Damage recognition in NER is tightly controlled by ubiquitin and SUMO modifications. Recent studies have shown that the SUMO-targeted ubiquitin ligase RNF111 promotes K63-linked ubiquitylation of SUMOylated XPC after DNA damage. However, the exact regulatory function of these modifications in vivo remains elusive. Here we show that RNF111 is required for efficient repair of ultraviolet-induced DNA lesions. RNF111-mediated ubiquitylation promotes the release of XPC from damaged DNA after NER initiation, and is needed for stable incorporation of the NER endonucleases XPG and ERCC1/XPF. Our data suggest that RNF111, together with the CRL4DDB2 ubiquitin ligase complex, is responsible for sequential XPC ubiquitylation, which regulates the recruitment and release of XPC and is crucial for efficient progression of the NER reaction, thereby providing an extra layer of quality control of NER.


Cell Cycle | 2014

ISWI chromatin remodeling complexes in the DNA damage response

Özge Z. Aydin; Wim Vermeulen; Hannes Lans

Regulation of chromatin structure is an essential component of the DNA damage response (DDR), which effectively preserves the integrity of DNA by a network of multiple DNA repair and associated signaling pathways. Within the DDR, chromatin is modified and remodeled to facilitate efficient DNA access, to control the activity of repair proteins and to mediate signaling. The mammalian ISWI family has recently emerged as one of the major ATP-dependent chromatin remodeling complex families that function in the DDR, as it is implicated in at least 3 major DNA repair pathways: homologous recombination, non-homologous end-joining and nucleotide excision repair. In this review, we discuss the various manners through which different ISWI complexes regulate DNA repair and how they are targeted to chromatin containing damaged DNA.


Nucleic Acids Research | 2014

Human ISWI complexes are targeted by SMARCA5 ATPase and SLIDE domains to help resolve lesion-stalled transcription.

Özge Z. Aydin; Jurgen A. Marteijn; Cristina Ribeiro-Silva; Aida Rodríguez López; Nils Wijgers; Godelieve Smeenk; Haico van Attikum; Raymond A. Poot; Wim Vermeulen; Hannes Lans

Chromatin compaction of deoxyribonucleic acid (DNA) presents a major challenge to the detection and removal of DNA damage. Helix-distorting DNA lesions that block transcription are specifically repaired by transcription-coupled nucleotide excision repair, which is initiated by binding of the CSB protein to lesion-stalled RNA polymerase II. Using live cell imaging, we identify a novel function for two distinct mammalian ISWI adenosine triphosphate (ATP)-dependent chromatin remodeling complexes in resolving lesion-stalled transcription. Human ISWI isoform SMARCA5/SNF2H and its binding partners ACF1 and WSTF are rapidly recruited to UV-C induced DNA damage to specifically facilitate CSB binding and to promote transcription recovery. SMARCA5 targeting to UV-C damage depends on transcription and histone modifications and requires functional SWI2/SNF2-ATPase and SLIDE domains. After initial recruitment to UV damage, SMARCA5 re-localizes away from the center of DNA damage, requiring its HAND domain. Our studies support a model in which SMARCA5 targeting to DNA damage-stalled transcription sites is controlled by an ATP-hydrolysis-dependent scanning and proofreading mechanism, highlighting how SWI2/SNF2 chromatin remodelers identify and bind nucleosomes containing damaged DNA.

Collaboration


Dive into the Hannes Lans's collaboration.

Top Co-Authors

Avatar

Wim Vermeulen

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Jurgen A. Marteijn

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Gert Jansen

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Jan H.J. Hoeijmakers

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arjan F. Theil

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mariangela Sabatella

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Özge Z. Aydin

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Christoffel Dinant

Erasmus University Rotterdam

View shared research outputs
Researchain Logo
Decentralizing Knowledge