Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 112022 10 21.
Article in English | MEDLINE | ID: mdl-36269050

ABSTRACT

The tumour suppressor PALB2 stimulates RAD51-mediated homologous recombination (HR) repair of DNA damage, whilst its steady-state association with active genes protects these loci from replication stress. Here, we report that the lysine acetyltransferases 2A and 2B (KAT2A/2B, also called GCN5/PCAF), two well-known transcriptional regulators, acetylate a cluster of seven lysine residues (7K-patch) within the PALB2 chromatin association motif (ChAM) and, in this way, regulate context-dependent PALB2 binding to chromatin. In unperturbed cells, the 7K-patch is targeted for KAT2A/2B-mediated acetylation, which in turn enhances the direct association of PALB2 with nucleosomes. Importantly, DNA damage triggers a rapid deacetylation of ChAM and increases the overall mobility of PALB2. Distinct missense mutations of the 7K-patch render the mode of PALB2 chromatin binding, making it either unstably chromatin-bound (7Q) or randomly bound with a reduced capacity for mobilisation (7R). Significantly, both of these mutations confer a deficiency in RAD51 foci formation and increase DNA damage in S phase, leading to the reduction of overall cell survival. Thus, our study reveals that acetylation of the ChAM 7K-patch acts as a molecular switch to enable dynamic PALB2 shuttling for HR repair while protecting active genes during DNA replication.


Subject(s)
Chromatin , Tumor Suppressor Proteins , Acetylation , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , DNA Repair , DNA Damage , Nucleosomes
2.
Oncotarget ; 8(39): 64918-64931, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29029401

ABSTRACT

The DNA damage response (DDR) is central to the cell survival and it requires post-translational modifications, in part, to sense the damage, amplify the signaling response and recruit and regulate DNA repair enzymes. Lysine methylation of histones such as H4K20 and non-histone proteins including p53 has been shown to be essential for the mounting of the DDR. It is well-known that the lysine methyltransferase SET7 regulates the DDR, as cells lacking this enzyme are hypersensitive to chemotherapeutic drugs. To define addition substrates of SET7 involved in the DDR, we screened a peptide array encompassing potential lysine methylation sites from >100 key DDR proteins and identified peptides from 58 proteins to be lysine methylated defining a methylation consensus sequence of [S>K-2; S>R-1; K0] consistent with previous findings. We focused on K377 methylation of the Flap endonuclease 1 (FEN1), a structure specific endonuclease with important functions in Okazaki fragment processing during DNA replication as a substrate of SET7. FEN1 was monomethylated by SET7 in vivo in a cell cycle dependent manner with levels increasing as cells progressed through S phase and decreasing as they exited S phase, as detected using K377me1 specific antibodies. Although K377me1 did not affect the enzymatic activity of FEN1, it was required for the cellular response to replicative stress by FEN1. These finding define FEN1 as a new substrate of SET7 required for the DDR.

3.
Proc Natl Acad Sci U S A ; 114(29): 7671-7676, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28673974

ABSTRACT

The partner and localiser of BRCA2 (PALB2) plays important roles in the maintenance of genome integrity and protection against cancer. Although PALB2 is commonly described as a repair factor recruited to sites of DNA breaks, recent studies provide evidence that PALB2 also associates with unperturbed chromatin. Here, we investigated the previously poorly described role of chromatin-associated PALB2 in undamaged cells. We found that PALB2 associates with active genes through its major binding partner, MRG15, which recognizes histone H3 trimethylated at lysine 36 (H3K36me3) by the SETD2 methyltransferase. Missense mutations that ablate PALB2 binding to MRG15 confer elevated sensitivity to the topoisomerase inhibitor camptothecin (CPT) and increased levels of aberrant metaphase chromosomes and DNA stress in gene bodies, which were suppressed by preventing DNA replication. Remarkably, the level of PALB2 at genic regions was frequently decreased, rather than increased, upon CPT treatment. We propose that the steady-state presence of PALB2 at active genes, mediated through the SETD2/H3K36me3/MRG15 axis, ensures an immediate response to DNA stress and therefore effective protection of these regions during DNA replication. This study provides a conceptual advance in demonstrating that the constitutive chromatin association of repair factors plays a key role in the maintenance of genome stability and furthers our understanding of why PALB2 defects lead to human genome instability syndromes.


Subject(s)
Chromatin/ultrastructure , DNA Damage , Fanconi Anemia Complementation Group N Protein/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Transcription Factors/metabolism , BRCA2 Protein/genetics , Cell Line, Tumor , Chromosomes/ultrastructure , DNA Repair , DNA Replication , Genome, Human , HEK293 Cells , HeLa Cells , Humans , Inhibitory Concentration 50 , Mutation , Protein Binding , Proteomics , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
4.
Nucleic Acids Res ; 45(14): 8341-8357, 2017 Aug 21.
Article in English | MEDLINE | ID: mdl-28666371

ABSTRACT

Fanconi anemia (FA) is a recessive genetic disorder characterized by congenital abnormalities, progressive bone-marrow failure, and cancer susceptibility. The FA pathway consists of at least 21 FANC genes (FANCA-FANCV), and the encoded protein products interact in a common cellular pathway to gain resistance against DNA interstrand crosslinks. After DNA damage, FANCD2 is monoubiquitinated and accumulates on chromatin. FANCD2 plays a central role in the FA pathway, using yet unidentified DNA binding regions. By using synthetic peptide mapping and DNA binding screen by electromobility shift assays, we found that FANCD2 bears two major DNA binding domains predominantly consisting of evolutionary conserved lysine residues. Furthermore, one domain at the N-terminus of FANCD2 bears also nuclear localization sequences for the protein. Mutations in the bifunctional DNA binding/NLS domain lead to a reduction in FANCD2 monoubiquitination and increase in mitomycin C sensitivity. Such phenotypes are not fully rescued by fusion with an heterologous NLS, which enable separation of DNA binding and nuclear import functions within this domain that are necessary for FANCD2 functions. Collectively, our results enlighten the importance of DNA binding and NLS residues in FANCD2 to activate an efficient FA pathway.


Subject(s)
DNA-Binding Proteins/genetics , DNA/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Nuclear Localization Signals/genetics , Amino Acid Sequence , Binding Sites/genetics , Cell Line, Tumor , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , DNA/metabolism , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Lysine/genetics , Lysine/metabolism , Microscopy, Fluorescence , Mutation , Protein Binding , RNA Interference , Signal Transduction/genetics , Ubiquitination
5.
Nucleic Acids Res ; 45(5): 2644-2657, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28158555

ABSTRACT

One typical mechanism to promote genomic instability, a hallmark of cancer, is to inactivate tumor suppressors, such as PALB2. It has recently been reported that mutations in PALB2 increase the risk of breast cancer by 8-9-fold by age 40 and the life time risk is ∼3-4-fold. To date, predicting the functional consequences of PALB2 mutations has been challenging as they lead to different cancer risks. Here, we performed a structure-function analysis of PALB2, using PALB2 truncated mutants (R170fs, L531fs, Q775X and W1038X), and uncovered a new mechanism by which cancer cells could drive genomic instability. Remarkably, the PALB2 W1038X mutant, harboring a mutation in its C-terminal domain, is still proficient in stimulating RAD51-mediated recombination in vitro, although it is unusually localized to the cytoplasm. After further investigation, we identified a hidden NES within the WD40 domain of PALB2 and found that the W1038X truncation leads to the exposure of this NES to CRM1, an export protein. This concept was also confirmed with another WD40-containing protein, RBBP4. Consequently, our studies reveal an unreported mechanism linking the nucleocytoplasmic translocation of PALB2 mutants to cancer formation.


Subject(s)
Mutation , Neoplasms/genetics , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Cytoplasm/metabolism , DNA/metabolism , Fanconi Anemia Complementation Group N Protein , HEK293 Cells , Humans , Karyopherins/metabolism , Nuclear Export Signals , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Rad51 Recombinase/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Deletion , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , WD40 Repeats , Exportin 1 Protein
6.
Elife ; 62017 02 27.
Article in English | MEDLINE | ID: mdl-28240985

ABSTRACT

DNA double-strand breaks (DSB) elicit a ubiquitylation cascade that controls DNA repair pathway choice. This cascade involves the ubiquitylation of histone H2A by the RNF168 ligase and the subsequent recruitment of RIF1, which suppresses homologous recombination (HR) in G1 cells. The RIF1-dependent suppression is relieved in S/G2 cells, allowing PALB2-driven HR to occur. With the inhibitory impact of RIF1 relieved, it remains unclear how RNF168-induced ubiquitylation influences HR. Here, we uncover that RNF168 links the HR machinery to H2A ubiquitylation in S/G2 cells. We show that PALB2 indirectly recognizes histone ubiquitylation by physically associating with ubiquitin-bound RNF168. This direct interaction is mediated by the newly identified PALB2-interacting domain (PID) in RNF168 and the WD40 domain in PALB2, and drives DNA repair by facilitating the assembly of PALB2-containing HR complexes at DSBs. Our findings demonstrate that RNF168 couples PALB2-dependent HR to H2A ubiquitylation to promote DNA repair and preserve genome integrity.


Subject(s)
DNA/metabolism , Fanconi Anemia Complementation Group N Protein/metabolism , Fibroblasts/metabolism , Histones/metabolism , Recombinational DNA Repair , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Line, Transformed , Cell Line, Tumor , DNA/genetics , DNA Breaks, Double-Stranded/radiation effects , Fanconi Anemia Complementation Group N Protein/genetics , Fibroblasts/cytology , Fibroblasts/radiation effects , HEK293 Cells , Histones/genetics , Humans , Lasers, Excimer , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/radiation effects , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoblasts/radiation effects , Protein Binding , Protein Interaction Domains and Motifs , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , X-Rays
7.
Nucleic Acids Res ; 44(22): 10879-10897, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27924011

ABSTRACT

APRIN (PDS5 cohesin associated factor B) interacts with both the cohesin complex and the BRCA2 tumor suppressor. How APRIN influences cohesion and DNA repair processes is not well understood. Here, we show that APRIN is recruited to DNA damage sites. We find that APRIN interacts directly with RAD51, PALB2 and BRCA2. APRIN stimulates RAD51-mediated DNA strand invasion. APRIN also binds DNA with an affinity for D-loop structures and single-strand (ss) DNA. APRIN is a new homologous recombination (HR) mediator as it counteracts the RPA inhibitory effect on RAD51 loading to ssDNA. We show that APRIN strongly improves the annealing of complementary-strand DNA and that it can stimulate this process in synergy with BRCA2. Unlike cohesin constituents, its depletion has no impact on class switch recombination, supporting a specific role for this protein in HR. Furthermore, we show that low APRIN expression levels correlate with a better survival in ovarian cancer patients and that APRIN depletion sensitizes cells to the PARP inhibitor Olaparib in xenografted zebrafish. Our findings establish APRIN as an important and specific actor of HR, with cohesin-independent functions.


Subject(s)
Biomarkers, Tumor/physiology , DNA-Binding Proteins/physiology , Ovarian Neoplasms/metabolism , Squamous Intraepithelial Lesions of the Cervix/metabolism , Transcription Factors/physiology , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , BRCA2 Protein/metabolism , Benzimidazoles/pharmacology , Biomarkers, Tumor/chemistry , Cell Line, Tumor , DNA Damage , DNA-Binding Proteins/chemistry , Drug Resistance, Neoplasm , Fanconi Anemia Complementation Group N Protein , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Nuclear Proteins/metabolism , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Phthalazines/pharmacology , Piperazines/pharmacology , Protein Binding , Protein Transport , ROC Curve , Rad51 Recombinase/metabolism , Recombinational DNA Repair , Squamous Intraepithelial Lesions of the Cervix/diagnosis , Squamous Intraepithelial Lesions of the Cervix/drug therapy , Squamous Intraepithelial Lesions of the Cervix/mortality , Transcription Factors/chemistry , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays , Zebrafish
8.
Biomolecules ; 5(4): 3204-59, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26610585

ABSTRACT

For decades, radiotherapy and chemotherapy were the two only approaches exploiting DNA repair processes to fight against cancer. Nowadays, cancer therapeutics can be a major challenge when it comes to seeking personalized targeted medicine that is both effective and selective to the malignancy. Over the last decade, the discovery of new targeted therapies against DNA damage signalling and repair has offered the possibility of therapeutic improvements in oncology. In this review, we summarize the current knowledge of DNA damage signalling and repair inhibitors, their molecular and cellular effects, and future therapeutic use.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage , DNA Repair Enzymes/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...