Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Cell ; 163(6): 1515-26, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26627737

ABSTRACT

The ability to perturb genes in human cells is crucial for elucidating gene function and holds great potential for finding therapeutic targets for diseases such as cancer. To extend the catalog of human core and context-dependent fitness genes, we have developed a high-complexity second-generation genome-scale CRISPR-Cas9 gRNA library and applied it to fitness screens in five human cell lines. Using an improved Bayesian analytical approach, we consistently discover 5-fold more fitness genes than were previously observed. We present a list of 1,580 human core fitness genes and describe their general properties. Moreover, we demonstrate that context-dependent fitness genes accurately recapitulate pathway-specific genetic vulnerabilities induced by known oncogenes and reveal cell-type-specific dependencies for specific receptor tyrosine kinases, even in oncogenic KRAS backgrounds. Thus, rigorous identification of human cell line fitness genes using a high-complexity CRISPR-Cas9 library affords a high-resolution view of the genetic vulnerabilities of a cell.


Subject(s)
Genes, Essential , Bayes Theorem , CRISPR-Cas Systems , Cell Line, Tumor , Gene Knockout Techniques , Gene Library , Humans , Mutation
2.
EMBO Rep ; 25(2): 725-744, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38177923

ABSTRACT

Viral infection often trigger an ATM serine/threonine kinase (ATM)-dependent DNA damage response in host cells that suppresses viral replication. Viruses evolved different strategies to counteract this antiviral surveillance system. Here, we report that human herpesvirus 6B (HHV-6B) infection causes genomic instability by suppressing ATM signaling in host cells. Expression of immediate-early protein 1 (IE1) phenocopies this phenotype and blocks homology-directed double-strand break repair. Mechanistically, IE1 interacts with NBS1, and inhibits ATM signaling through two distinct domains. HHV-6B seems to efficiently inhibit ATM signaling as further depletion of either NBS1 or ATM do not significantly boost viral replication in infected cells. Interestingly, viral integration of HHV-6B into the host's telomeres is not strictly dependent on NBS1, challenging current models where integration occurs through homology-directed repair. Given that spontaneous IE1 expression has been detected in cells of subjects with inherited chromosomally-integrated form of HHV-6B (iciHHV-6B), a condition associated with several health conditions, our results raise the possibility of a link between genomic instability and the development of iciHHV-6-associated diseases.


Subject(s)
Herpesvirus 6, Human , Immediate-Early Proteins , Roseolovirus Infections , Humans , Herpesvirus 6, Human/genetics , Herpesvirus 6, Human/metabolism , Roseolovirus Infections/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Virus Integration , Genomic Instability , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism
3.
Mol Cell ; 62(3): 409-421, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27153538

ABSTRACT

The NuA4/TIP60 acetyltransferase complex is a key regulator of genome expression and stability. Here we 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. We find 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.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Histone Acetyltransferases/metabolism , Histones/metabolism , Protein Processing, Post-Translational , Tumor Suppressor p53-Binding Protein 1/metabolism , Acetylation , Binding Sites , Binding, Competitive , CRISPR-Cas Systems , Chromosomal Proteins, Non-Histone/genetics , DNA Breaks, Double-Stranded , DNA Repair , Histone Acetyltransferases/genetics , Histones/genetics , Humans , K562 Cells , Lysine Acetyltransferase 5 , Promoter Regions, Genetic , Protein Binding , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Time Factors , Transcription, Genetic , Transfection , Tumor Suppressor p53-Binding Protein 1/genetics , Ubiquitination
4.
EMBO Rep ; 22(12): e53679, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34726323

ABSTRACT

The tumor suppressor BRCA1 accumulates at sites of DNA damage in a ubiquitin-dependent manner. In this work, we revisit the role of RAP80 in promoting BRCA1 recruitment to damaged chromatin. We find that RAP80 acts redundantly with the BRCA1 RING domain to promote BRCA1 recruitment to DNA damage sites. We show that that RNF8 E3 ligase acts upstream of both the RAP80- and RING-dependent activities, whereas RNF168 acts uniquely upstream of the RING domain. BRCA1 RING mutations that do not impact BARD1 interaction, such as the E2 binding-deficient I26A mutation, render BRCA1 unable to accumulate at DNA damage sites in the absence of RAP80. Cells that combine BRCA1 I26A and mutations that disable the RAP80-BRCA1 interaction are hypersensitive to PARP inhibition and are unable to form RAD51 foci. Our results suggest that in the absence of RAP80, the BRCA1 E3 ligase activity is necessary for recognition of histone H2A Lys13/Lys15 ubiquitylation by BARD1, although we cannot rule out the possibility that the BRCA1 RING facilitates ubiquitylated nucleosome recognition in other ways.


Subject(s)
Nuclear Proteins , Ubiquitin , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Carrier Proteins/metabolism , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Histone Chaperones/genetics , Histone Chaperones/metabolism , Nuclear Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Nature ; 536(7614): 100-3, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27462807

ABSTRACT

DNA double-strand breaks (DSBs) elicit a histone modification cascade that controls DNA repair. This pathway involves the sequential ubiquitination of histones H1 and H2A by the E3 ubiquitin ligases RNF8 and RNF168, respectively. RNF168 ubiquitinates H2A on lysine 13 and lysine 15 (refs 7, 8) (yielding H2AK13ub and H2AK15ub, respectively), an event that triggers the recruitment of 53BP1 (also known as TP53BP1) to chromatin flanking DSBs. 53BP1 binds specifically to H2AK15ub-containing nucleosomes through a peptide segment termed the ubiquitination-dependent recruitment motif (UDR), which requires the simultaneous engagement of histone H4 lysine 20 dimethylation (H4K20me2) by its tandem Tudor domain. How 53BP1 interacts with these two histone marks in the nucleosomal context, how it recognizes ubiquitin, and how it discriminates between H2AK13ub and H2AK15ub is unknown. Here we present the electron cryomicroscopy (cryo-EM) structure of a dimerized human 53BP1 fragment bound to a H4K20me2-containing and H2AK15ub-containing nucleosome core particle (NCP-ubme) at 4.5 Å resolution. The structure reveals that H4K20me2 and H2AK15ub recognition involves intimate contacts with multiple nucleosomal elements including the acidic patch. Ubiquitin recognition by 53BP1 is unusual and involves the sandwiching of the UDR segment between ubiquitin and the NCP surface. The selectivity for H2AK15ub is imparted by two arginine fingers in the H2A amino-terminal tail, which straddle the nucleosomal DNA and serve to position ubiquitin over the NCP-bound UDR segment. The structure of the complex between NCP-ubme and 53BP1 reveals the basis of 53BP1 recruitment to DSB sites and illuminates how combinations of histone marks and nucleosomal elements cooperate to produce highly specific chromatin responses, such as those elicited following chromosome breaks.


Subject(s)
Cryoelectron Microscopy , Histones/chemistry , Histones/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Nucleosomes/metabolism , Nucleosomes/ultrastructure , DNA Breaks, Double-Stranded , DNA Repair , Humans , Methylation , Models, Molecular , Nucleosomes/chemistry , Nucleosomes/genetics , Pliability , Protein Multimerization , Protein Structure, Tertiary , Substrate Specificity , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin/metabolism , Ubiquitination
6.
Proc Natl Acad Sci U S A ; 116(39): 19552-19562, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31501315

ABSTRACT

High-risk human papillomaviruses (HR-HPVs) promote cervical cancer as well as a subset of anogenital and head and neck cancers. Due to their limited coding capacity, HPVs hijack the host cell's DNA replication and repair machineries to replicate their own genomes. How this host-pathogen interaction contributes to genomic instability is unknown. Here, we report that HPV-infected cancer cells express high levels of RNF168, an E3 ubiquitin ligase that is critical for proper DNA repair following DNA double-strand breaks, and accumulate high numbers of 53BP1 nuclear bodies, a marker of genomic instability induced by replication stress. We describe a mechanism by which HPV E7 subverts the function of RNF168 at DNA double-strand breaks, providing a rationale for increased homology-directed recombination in E6/E7-expressing cervical cancer cells. By targeting a new regulatory domain of RNF168, E7 binds directly to the E3 ligase without affecting its enzymatic activity. As RNF168 knockdown impairs viral genome amplification in differentiated keratinocytes, we propose that E7 hijacks the E3 ligase to promote the viral replicative cycle. This study reveals a mechanism by which tumor viruses reshape the cellular response to DNA damage by manipulating RNF168-dependent ubiquitin signaling. Importantly, our findings reveal a pathway by which HPV may promote the genomic instability that drives oncogenesis.


Subject(s)
DNA Breaks, Double-Stranded , Papillomaviridae/metabolism , Papillomavirus E7 Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , DNA Repair , Female , Genomic Instability , Homologous Recombination , Host-Pathogen Interactions , Humans , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Signal Transduction , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics , Uterine Cervical Neoplasms/virology
7.
Mol Cell ; 49(5): 872-83, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23333306

ABSTRACT

DNA double-strand break (DSB) repair pathway choice is governed by the opposing activities of 53BP1 and BRCA1. 53BP1 stimulates nonhomologous end joining (NHEJ), whereas BRCA1 promotes end resection and homologous recombination (HR). Here we show that 53BP1 is an inhibitor of BRCA1 accumulation at DSB sites, specifically in the G1 phase of the cell cycle. ATM-dependent phosphorylation of 53BP1 physically recruits RIF1 to DSB sites, and we identify RIF1 as the critical effector of 53BP1 during DSB repair. Remarkably, RIF1 accumulation at DSB sites is strongly antagonized by BRCA1 and its interacting partner CtIP. Lastly, we show that depletion of RIF1 is able to restore end resection and RAD51 loading in BRCA1-depleted cells. This work therefore identifies a cell cycle-regulated circuit, underpinned by RIF1 and BRCA1, that governs DSB repair pathway choice to ensure that NHEJ dominates in G1 and HR is favored from S phase onward.


Subject(s)
BRCA1 Protein/genetics , Carrier Proteins/genetics , Cell Cycle/genetics , DNA Repair , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Telomere-Binding Proteins/genetics , BRCA1 Protein/metabolism , Binding Sites , Carrier Proteins/metabolism , DNA End-Joining Repair/genetics , Endodeoxyribonucleases , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , S Phase , Telomere-Binding Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1
8.
J Virol ; 93(22)2019 11 15.
Article in English | MEDLINE | ID: mdl-31462557

ABSTRACT

The BMRF1 protein of Epstein-Barr virus (EBV) has multiple roles in viral lytic infection, including serving as the DNA polymerase processivity factor, activating transcription from several EBV promoters and inhibiting the host DNA damage response to double-stranded DNA breaks (DSBs). Using affinity purification coupled to mass spectrometry, we identified the nucleosome remodeling and deacetylation (NuRD) complex as the top interactor of BMRF1. We further found that NuRD components localize with BMRF1 at viral replication compartments and that this interaction occurs through the BMRF1 C-terminal region previously shown to mediate transcriptional activation. We identified an RBBP4 binding motif within this region that can interact with both RBBP4 and MTA2 components of the NuRD complex and showed that point mutation of this motif abrogates NuRD binding as well as the ability of BMRF1 to activate transcription from the BDLF3 and BLLF1 EBV promoters. In addition to its role in transcriptional regulation, NuRD has been shown to contribute to DSB signaling in enabling recruitment of RNF168 ubiquitin ligase and subsequent ubiquitylation at the break. We showed that BMRF1 inhibited RNF168 recruitment and ubiquitylation at DSBs and that this inhibition was at least partly relieved by loss of the NuRD interaction. The results reveal a mechanism by which BMRF1 activates transcription and inhibits DSB signaling and a novel role for NuRD in transcriptional activation in EBV.IMPORTANCE The Epstein-Barr virus (EBV) BMRF1 protein is critical for EBV infection, playing key roles in viral genome replication, activation of EBV genes, and inhibition of host DNA damage responses (DDRs). Here we show that BMRF1 targets the cellular nucleosome remodeling and deacetylation (NuRD) complex, using a motif in the BMRF1 transcriptional activation sequence. Mutation of this motif disrupts the ability of BMRF1 to activate transcription and interfere with DDRs, showing the importance of the NuRD interaction for BMRF1 functions. BMRF1 was shown to act at the same step in the DDR as NuRD, suggesting that it interferes with NuRD function.


Subject(s)
Antigens, Viral/metabolism , DNA Damage , Herpesvirus 4, Human/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Antigens, Viral/genetics , Cell Line, Tumor , DNA Replication , DNA, Viral/genetics , DNA-Binding Proteins/metabolism , Epstein-Barr Virus Infections/virology , HEK293 Cells , HeLa Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Membrane Glycoproteins/metabolism , Promoter Regions, Genetic , Signal Transduction , Trans-Activators/metabolism , Transcriptional Activation , Viral Proteins/metabolism , Virus Replication
9.
Mol Cell ; 47(3): 383-95, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22742833

ABSTRACT

The response to DNA double-strand breaks (DSBs) entails the hierarchical recruitment of proteins orchestrated by ATM-dependent phosphorylation and RNF8-mediated chromatin ubiquitylation. As in most ubiquitin-dependent processes, the ordered accumulation of DNA repair factors at the break site relies on ubiquitin-binding domains (UBDs). However, how UBDs select their ligands is poorly understood, and therefore we sought to uncover the basis for selectivity in the ubiquitin-dependent DSB response. We show that RNF168, its paralog RNF169, RAD18, and the BRCA1-interacting RAP80 protein accumulate at DSB sites through the use of bipartite modules composed of UBDs juxtaposed to peptide motifs that provide specificity. These sequences, named LR motifs (LRMs), are transferable, and we show that the RNF169 LRM2 binds to nucleosomes, the substrates of RNF168. The LRM-based selection of ligands is a parsimonious means to build a highly discrete ubiquitin-based signaling pathway such as the DNA damage response.


Subject(s)
Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HCT116 Cells , HEK293 Cells , Histone Chaperones , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphorylation/physiology , Ubiquitin/physiology , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination/physiology
10.
Nucleic Acids Res ; 46(D1): D901-D910, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29126202

ABSTRACT

Interpretation of genetic variation is needed for deciphering genotype-phenotype associations, mechanisms of inherited disease, and cancer driver mutations. Millions of single nucleotide variants (SNVs) in human genomes are known and thousands are associated with disease. An estimated 21% of disease-associated amino acid substitutions corresponding to missense SNVs are located in protein sites of post-translational modifications (PTMs), chemical modifications of amino acids that extend protein function. ActiveDriverDB is a comprehensive human proteo-genomics database that annotates disease mutations and population variants through the lens of PTMs. We integrated >385,000 published PTM sites with ∼3.6 million substitutions from The Cancer Genome Atlas (TCGA), the ClinVar database of disease genes, and human genome sequencing projects. The database includes site-specific interaction networks of proteins, upstream enzymes such as kinases, and drugs targeting these enzymes. We also predicted network-rewiring impact of mutations by analyzing gains and losses of kinase-bound sequence motifs. ActiveDriverDB provides detailed visualization, filtering, browsing and searching options for studying PTM-associated mutations. Users can upload mutation datasets interactively and use our application programming interface in pipelines. Integrative analysis of mutations and PTMs may help decipher molecular mechanisms of phenotypes and disease, as exemplified by case studies of TP53, BRCA2 and VHL. The open-source database is available at https://www.ActiveDriverDB.org.


Subject(s)
Databases, Genetic , Databases, Protein , Disease/genetics , Mutation , Protein Processing, Post-Translational/genetics , Amino Acid Substitution , Data Mining/methods , Datasets as Topic , Genetic Association Studies , Genetic Variation , Genome, Human , Genomics , Humans , Molecular Sequence Annotation , Polymorphism, Single Nucleotide , Protein Kinases/genetics , Proteomics , Software , User-Computer Interface
11.
J Virol ; 92(14)2018 07 15.
Article in English | MEDLINE | ID: mdl-29743367

ABSTRACT

To replicate and persist in human cells, linear double-stranded DNA (dsDNA) viruses, such as Epstein-Barr virus (EBV), must overcome the host DNA damage response (DDR) that is triggered by the viral genomes. Since this response is necessary to maintain cellular genome integrity, its inhibition by EBV is likely an important factor in the development of cancers associated with EBV infection, including gastric carcinoma. Here we present the first extensive screen of EBV proteins that inhibit dsDNA break signaling. We identify the BKRF4 tegument protein as a DDR inhibitor that interferes with histone ubiquitylation at dsDNA breaks and recruitment of the RNF168 histone ubiquitin ligase. We further show that BKRF4 binds directly to histones through an acidic domain that targets BKRF4 to cellular chromatin and is sufficient to inhibit dsDNA break signaling. BKRF4 transcripts were detected in EBV-positive gastric carcinoma cells (AGS-EBV), and these increased in lytic infection. Silencing of BKRF4 in both latent and lytic AGS-EBV cells (but not in EBV-negative AGS cells) resulted in increased dsDNA break signaling, confirming a role for BKRF4 in DDR inhibition in the context of EBV infection and suggesting that BKRF4 is expressed in latent cells. BKRF4 was also found to be consistently expressed in EBV-positive gastric tumors in the absence of a full lytic infection. The results suggest that BKRF4 plays a role in inhibiting the cellular DDR in latent and lytic EBV infection and that the resulting accumulation of DNA damage might contribute to development of gastric carcinoma.IMPORTANCE Epstein-Barr virus (EBV) infects most people worldwide and is causatively associated with several types of cancer, including ∼10% of gastric carcinomas. EBV encodes ∼80 proteins, many of which are believed to manipulate cellular regulatory pathways but are poorly characterized. The DNA damage response (DDR) is one such pathway that is critical for maintaining genome integrity and preventing cancer-associated mutations. In this study, a screen for EBV proteins that inhibit the DDR identified BKRF4 as a DDR inhibitor that binds histones and blocks their ubiquitylation at the DNA damage sites. We also present evidence that BKRF4 is expressed in both latent and lytic forms of EBV infection, where it downregulates the DDR, as well as in EBV-positive gastric tumors. The results suggest that BKRF4 could contribute to the development of gastric carcinoma through its ability to inhibit the DDR.


Subject(s)
Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/physiology , Histones/metabolism , Stomach Neoplasms/virology , Viral Proteins/genetics , Viral Proteins/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair , Epstein-Barr Virus Infections/genetics , Gene Expression Regulation, Viral , Gene Library , HEK293 Cells , Humans , Protein Domains , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Viral Proteins/chemistry , Virus Replication
12.
Nature ; 499(7456): 50-4, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23760478

ABSTRACT

53BP1 (also called TP53BP1) is a chromatin-associated factor that promotes immunoglobulin class switching and DNA double-strand-break (DSB) repair by non-homologous end joining. To accomplish its function in DNA repair, 53BP1 accumulates at DSB sites downstream of the RNF168 ubiquitin ligase. How ubiquitin recruits 53BP1 to break sites remains unknown as its relocalization involves recognition of histone H4 Lys 20 (H4K20) methylation by its Tudor domain. Here we elucidate how vertebrate 53BP1 is recruited to the chromatin that flanks DSB sites. We show that 53BP1 recognizes mononucleosomes containing dimethylated H4K20 (H4K20me2) and H2A ubiquitinated on Lys 15 (H2AK15ub), the latter being a product of RNF168 action on chromatin. 53BP1 binds to nucleosomes minimally as a dimer using its previously characterized methyl-lysine-binding Tudor domain and a carboxy-terminal extension, termed the ubiquitination-dependent recruitment (UDR) motif, which interacts with the epitope formed by H2AK15ub and its surrounding residues on the H2A tail. 53BP1 is therefore a bivalent histone modification reader that recognizes a histone 'code' produced by DSB signalling.


Subject(s)
DNA Damage , Histones/chemistry , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/metabolism , Ubiquitin/metabolism , Ubiquitination , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Line , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleosomes/chemistry , Nucleosomes/metabolism , Protein Binding , Protein Structure, Tertiary , Schizosaccharomyces , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Signal Transduction , Tumor Suppressor p53-Binding Protein 1
13.
Proc Natl Acad Sci U S A ; 110(52): 20982-7, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24324146

ABSTRACT

Defective signaling or repair of DNA double-strand breaks has been associated with developmental defects and human diseases. The E3 ligase RING finger 168 (RNF168), mutated in the human radiosensitivity, immunodeficiency, dysmorphic features, and learning difficulties syndrome, was shown to ubiquitylate H2A-type histones, and this ubiquitylation was proposed to facilitate the recruitment of p53-binding protein 1 (53BP1) to the sites of DNA double-strand breaks. In contrast to more upstream proteins signaling DNA double-strand breaks (e.g., RNF8), deficiency of RNF168 fully prevents both the initial recruitment to and retention of 53BP1 at sites of DNA damage; however, the mechanism for this difference has remained unclear. Here, we identify mechanisms that regulate 53BP1 recruitment to the sites of DNA double-strand breaks and provide evidence that RNF168 plays a central role in the regulation of 53BP1 functions. RNF168 mediates K63-linked ubiquitylation of 53BP1 which is required for the initial recruitment of 53BP1 to sites of DNA double-strand breaks and for its function in DNA damage repair, checkpoint activation, and genomic integrity. Our findings highlight the multistep roles of RNF168 in signaling DNA damage.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Animals , DNA Repair/genetics , Fibroblasts , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/physiology , Mice , Tumor Suppressor p53-Binding Protein 1
14.
J Virol ; 86(7): 3486-500, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22278251

ABSTRACT

The papillomavirus E1 helicase is recruited by E2 to the viral origin, where it assembles into a double hexamer that orchestrates replication of the viral genome. We previously identified the cellular WD40 repeat-containing protein p80/UAF1 as a novel interaction partner of E1 from anogenital human papillomavirus (HPV) types. p80 was found to interact with the first 40 residues of HPV type 31 (HPV31) E1, and amino acid substitutions within this domain abrogated the maintenance of the viral episome in keratinocytes. In this study, we report that these p80-binding substitutions reduce by 70% the ability of E1 to support transient viral DNA replication without affecting its interaction with E2 and assembly at the origin in vivo. Microscopy studies revealed that p80 is relocalized from the cytoplasm to discrete subnuclear foci by E1 and E2. Chromatin immunoprecipitation assays further revealed that p80 is recruited to the viral origin in an E1- and E2-dependent manner. Interestingly, overexpression of a 40-amino-acid-long p80-binding peptide, derived from HPV31 E1, was found to inhibit viral DNA replication by preventing the recruitment of endogenous p80 to the origin. Mutant peptides defective for p80 interaction were not inhibitory, demonstrating the specificity of this effect. Characterization of this E1 peptide by nuclear magnetic resonance (NMR) showed that it is intrinsically disordered in solution, while mapping studies indicated that the WD repeats of p80 are required for E1 interaction. These results provide additional evidence for the requirement for p80 in anogenital HPV DNA replication and highlight the potential of E1-p80 interaction as a novel antiviral target.


Subject(s)
DNA Replication , Down-Regulation , Human papillomavirus 31/genetics , Papillomavirus Infections/metabolism , Peptides/metabolism , Proteins/metabolism , Viral Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Human papillomavirus 31/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Peptides/genetics , Protein Binding , Protein Structure, Tertiary , Proteins/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
15.
Life Sci Alliance ; 6(6)2023 06.
Article in English | MEDLINE | ID: mdl-36882285

ABSTRACT

Spinal muscular atrophy is the leading genetic cause of infant mortality and results from depleted levels of functional survival of motor neuron (SMN) protein by either deletion or mutation of the SMN1 gene. SMN is characterized by a central TUDOR domain, which mediates the association of SMN with arginine methylated (Rme) partners, such as coilin, fibrillarin, and RNA pol II (RNA polymerase II). Herein, we biochemically demonstrate that SMN also associates with histone H3 monomethylated on lysine 79 (H3K79me1), defining SMN as not only the first protein known to associate with the H3K79me1 histone modification but also the first histone mark reader to recognize both methylated arginine and lysine residues. Mutational analyzes provide evidence that SMNTUDOR associates with H3 via an aromatic cage. Importantly, most SMNTUDOR mutants found in spinal muscular atrophy patients fail to associate with H3K79me1.


Subject(s)
Histone Code , Muscular Atrophy, Spinal , Survival of Motor Neuron 1 Protein , Humans , Infant , Arginine , Lysine , Muscular Atrophy, Spinal/genetics , RNA Polymerase II , Transcription Factors , Survival of Motor Neuron 1 Protein/genetics
16.
Nat Commun ; 14(1): 381, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36693839

ABSTRACT

Fanconi Anemia (FA) is a rare, genome instability-associated disease characterized by a deficiency in repairing DNA crosslinks, which are known to perturb several cellular processes, including DNA transcription, replication, and repair. Formaldehyde, a by-product of metabolism, is thought to drive FA by generating DNA interstrand crosslinks (ICLs) and DNA-protein crosslinks (DPCs). However, the impact of formaldehyde on global cellular pathways has not been investigated thoroughly. Herein, using a pangenomic CRISPR-Cas9 screen, we identify EXO1 as a critical regulator of formaldehyde-induced DNA lesions. We show that EXO1 knockout cell lines exhibit formaldehyde sensitivity leading to the accumulation of replicative stress, DNA double-strand breaks, and quadriradial chromosomes, a typical feature of FA. After formaldehyde exposure, EXO1 is recruited to chromatin, protects DNA replication forks from degradation, and functions in parallel with the FA pathway to promote cell survival. In vitro, EXO1-mediated exonuclease activity is proficient in removing DPCs. Collectively, we show that EXO1 limits replication stress and DNA damage to counteract formaldehyde-induced genome instability.


Subject(s)
CRISPR-Cas Systems , Drug Tolerance , Exodeoxyribonucleases , Fanconi Anemia , Formaldehyde , Humans , DNA , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair/drug effects , DNA Repair/genetics , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA Replication/drug effects , DNA Replication/genetics , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Fanconi Anemia/chemically induced , Fanconi Anemia/genetics , Formaldehyde/toxicity , Genomic Instability/drug effects , Genomic Instability/genetics , Drug Tolerance/genetics
17.
Sci Adv ; 9(32): eadf4082, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37556550

ABSTRACT

Interstrand DNA cross-links (ICLs) represent complex lesions that compromise genomic stability. Several pathways have been involved in ICL repair, but the extent of factors involved in the resolution of ICL-induced DNA double-strand breaks (DSBs) remains poorly defined. Using CRISPR-based genomics, we identified FIGNL1 interacting regulator of recombination and mitosis (FIRRM) as a sensitizer of the ICL-inducing agent mafosfamide. Mechanistically, we showed that FIRRM, like its interactor Fidgetin like 1 (FIGNL1), contributes to the resolution of RAD51 foci at ICL-induced DSBs. While the stability of FIGNL1 and FIRRM is interdependent, expression of a mutant of FIRRM (∆WCF), which stabilizes the protein in the absence of FIGNL1, allows the resolution of RAD51 foci and cell survival, suggesting that FIRRM has FIGNL1-independent function during DNA repair. In line with this model, FIRRM binds preferentially single-stranded DNA in vitro, raising the possibility that it directly contributes to RAD51 disassembly by interacting with DNA. Together, our findings establish FIRRM as a promoting factor of ICL repair.


Subject(s)
DNA Repair , Rad51 Recombinase , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Proteins/genetics , DNA/genetics , Mitosis
18.
Nat Commun ; 14(1): 697, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36754959

ABSTRACT

Human acetyltransferases MOZ and MORF are implicated in chromosomal translocations associated with aggressive leukemias. Oncogenic translocations involve the far amino terminus of MOZ/MORF, the function of which remains unclear. Here, we identified and characterized two structured winged helix (WH) domains, WH1 and WH2, in MORF and MOZ. WHs bind DNA in a cooperative manner, with WH1 specifically recognizing unmethylated CpG sequences. Structural and genomic analyses show that the DNA binding function of WHs targets MORF/MOZ to gene promoters, stimulating transcription and H3K23 acetylation, and WH1 recruits oncogenic fusions to HOXA genes that trigger leukemogenesis. Cryo-EM, NMR, mass spectrometry and mutagenesis studies provide mechanistic insight into the DNA-binding mechanism, which includes the association of WH1 with the CpG-containing linker DNA and binding of WH2 to the dyad of the nucleosome. The discovery of WHs in MORF and MOZ and their DNA binding functions could open an avenue in developing therapeutics to treat diseases associated with aberrant MOZ/MORF acetyltransferase activities.


Subject(s)
Acetyltransferases , Histone Acetyltransferases , Leukemia , Humans , Acetylation , Acetyltransferases/metabolism , CpG Islands/genetics , Histone Acetyltransferases/metabolism , Leukemia/genetics , Translocation, Genetic
19.
Cell Rep ; 42(7): 112668, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37347663

ABSTRACT

Joint DNA molecules are natural byproducts of DNA replication and repair. Persistent joint molecules give rise to ultrafine DNA bridges (UFBs) in mitosis, compromising sister chromatid separation. The DNA translocase PICH (ERCC6L) has a central role in UFB resolution. A genome-wide loss-of-function screen is performed to identify the genetic context of PICH dependency. In addition to genes involved in DNA condensation, centromere stability, and DNA-damage repair, we identify FIGNL1-interacting regulator of recombination and mitosis (FIRRM), formerly known as C1orf112. We find that FIRRM interacts with and stabilizes the AAA+ ATPase FIGNL1. Inactivation of either FIRRM or FIGNL1 results in UFB formation, prolonged accumulation of RAD51 at nuclear foci, and impaired replication fork dynamics and consequently impairs genome maintenance. Combined, our data suggest that inactivation of FIRRM and FIGNL1 dysregulates RAD51 dynamics at replication forks, resulting in persistent DNA lesions and a dependency on PICH to preserve cell viability.


Subject(s)
Mitosis , Proteins , Proteins/genetics , Adenosine Triphosphatases/metabolism , DNA , Chromatids/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , DNA Replication/genetics , DNA Damage
20.
J Virol ; 85(2): 818-27, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980496

ABSTRACT

The origin-binding domain (OBD) of simian virus 40 (SV40) large T-antigen (T-Ag) is essential for many of T-Ag's interactions with DNA. Nevertheless, many important issues related to DNA binding, for example, how single-stranded DNA (ssDNA) transits along the T-Ag OBD, have yet to be established. Therefore, X-ray crystallography was used to determine the costructure of the T-Ag OBD bound to DNA substrates such as the single-stranded region of a forked oligonucleotide. A second structure of the T-Ag OBD crystallized in the presence of poly(dT)(12) is also reported. To test the conclusions derived from these structures, residues identified as being involved in binding to ssDNA by crystallography or by an earlier nuclear magnetic resonance study were mutated, and their binding to DNA was characterized via fluorescence anisotropy. In addition, these mutations were introduced into full-length T-Ag, and these mutants were tested for their ability to support replication. When considered in terms of additional homology-based sequence alignments, our studies refine our understanding of how the T-Ag OBDs encoded by the polyomavirus family interact with ssDNA, a critical step during the initiation of DNA replication.


Subject(s)
Antigens, Viral, Tumor/chemistry , Antigens, Viral, Tumor/metabolism , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , Simian virus 40/physiology , Amino Acid Sequence , Animals , Antigens, Viral, Tumor/genetics , Crystallography, X-Ray , Fluorescence Polarization , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL