Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Nucleic Acids Res ; 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38828775

The clinical success of PARP1/2 inhibitors (PARPi) prompts the expansion of their applicability beyond homologous recombination deficiency. Here, we demonstrate that the loss of the accessory subunits of DNA polymerase epsilon, POLE3 and POLE4, sensitizes cells to PARPi. We show that the sensitivity of POLE4 knockouts is not due to compromised response to DNA damage or homologous recombination deficiency. Instead, POLE4 loss affects replication speed leading to the accumulation of single-stranded DNA gaps behind replication forks upon PARPi treatment, due to impaired post-replicative repair. POLE4 knockouts elicit elevated replication stress signaling involving ATR and DNA-PK. We find POLE4 to act parallel to BRCA1 in inducing sensitivity to PARPi and counteracts acquired resistance associated with restoration of homologous recombination. Altogether, our findings establish POLE4 as a promising target to improve PARPi driven therapies and hamper acquired PARPi resistance.

2.
Mol Biol Cell ; 35(3): br7, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38170578

ADP-ribosylation signaling orchestrates the recruitment of various repair actors and chromatin remodeling processes promoting access to lesions during the early stages of the DNA damage response. The chromatin remodeler complex ACF, composed of the ATPase subunit SMARCA5/SNF2H and the cofactor ACF1/BAZ1A, is among the factors that accumulate at DNA lesions in an ADP-ribosylation dependent manner. In this work, we show that each subunit of the ACF complex accumulates to DNA breaks independently from its partner. Furthermore, we demonstrate that the recruitment of SMARCA5 and ACF1 to sites of damage is not due to direct binding to the ADP-ribose moieties but due to facilitated DNA binding at relaxed ADP-ribosylated chromatin. Therefore, our work provides new insights regarding the mechanisms underlying the timely accumulation of ACF1 and SMARCA5 to DNA lesions, where they contribute to efficient DNA damage resolution.


Chromatin , DNA Damage , DNA/metabolism , Adenosine Triphosphatases/metabolism , DNA Repair , ADP-Ribosylation
3.
Cell Rep ; 42(9): 113113, 2023 09 26.
Article En | MEDLINE | ID: mdl-37676774

The timely removal of ADP-ribosylation is crucial for efficient DNA repair. However, much remains to be discovered about ADP-ribosylhydrolases. Here, we characterize the physiological role of TARG1, an ADP-ribosylhydrolase that removes aspartate/glutamate-linked ADP-ribosylation. We reveal its function in the DNA damage response and show that the loss of TARG1 sensitizes cells to inhibitors of topoisomerase II, ATR, and PARP. Furthermore, we find a PARP1-mediated synthetic lethal interaction between TARG1 and PARG, driven by the toxic accumulation of ADP-ribosylation, that induces replication stress and genomic instability. Finally, we show that histone PARylation factor 1 (HPF1) deficiency exacerbates the toxicity and genomic instability induced by excessive ADP-ribosylation, suggesting a close crosstalk between components of the serine- and aspartate/glutamate-linked ADP-ribosylation pathways. Altogether, our data identify TARG1 as a potential biomarker for the response of cancer cells to PARP and PARG inhibition and establish that the interplay of TARG1 and PARG protects cells against genomic instability.


Aspartic Acid , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Aspartic Acid/metabolism , ADP-Ribosylation , Genomic Instability , Glutamates/metabolism , Carrier Proteins/metabolism , Nuclear Proteins/metabolism
4.
Mol Cell ; 83(10): 1743-1760.e11, 2023 05 18.
Article En | MEDLINE | ID: mdl-37116497

PARP1, an established anti-cancer target that regulates many cellular pathways, including DNA repair signaling, has been intensely studied for decades as a poly(ADP-ribosyl)transferase. Although recent studies have revealed the prevalence of mono-ADP-ribosylation upon DNA damage, it was unknown whether this signal plays an active role in the cell or is just a byproduct of poly-ADP-ribosylation. By engineering SpyTag-based modular antibodies for sensitive and flexible detection of mono-ADP-ribosylation, including fluorescence-based sensors for live-cell imaging, we demonstrate that serine mono-ADP-ribosylation constitutes a second wave of PARP1 signaling shaped by the cellular HPF1/PARP1 ratio. Multilevel chromatin proteomics reveals histone mono-ADP-ribosylation readers, including RNF114, a ubiquitin ligase recruited to DNA lesions through a zinc-finger domain, modulating the DNA damage response and telomere maintenance. Our work provides a technological framework for illuminating ADP-ribosylation in a wide range of applications and biological contexts and establishes mono-ADP-ribosylation by HPF1/PARP1 as an important information carrier for cell signaling.


ADP-Ribosylation , Histones , Histones/genetics , Histones/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Chromatin , DNA Damage , Antibodies/genetics , Signal Transduction
5.
Nat Struct Mol Biol ; 30(5): 678-691, 2023 05.
Article En | MEDLINE | ID: mdl-37106138

Poly(ADP-ribose) polymerase 1 (PARP1) activity is regulated by its co-factor histone poly(ADP-ribosylation) factor 1 (HPF1). The complex formed by HPF1 and PARP1 catalyzes ADP-ribosylation of serine residues of proteins near DNA breaks, mainly PARP1 and histones. However, the effect of HPF1 on DNA repair regulated by PARP1 remains unclear. Here, we show that HPF1 controls prolonged histone ADP-ribosylation in the vicinity of the DNA breaks by regulating both the number and length of ADP-ribose chains. Furthermore, we demonstrate that HPF1-dependent histone ADP-ribosylation triggers the rapid unfolding of chromatin, facilitating access to DNA at sites of damage. This process promotes the assembly of both the homologous recombination and non-homologous end joining repair machineries. Altogether, our data highlight the key roles played by the PARP1/HPF1 complex in regulating ADP-ribosylation signaling as well as the conformation of damaged chromatin at early stages of the DNA damage response.


Chromatin , Histones , Histones/metabolism , Poly(ADP-ribose) Polymerases/chemistry , Poly (ADP-Ribose) Polymerase-1/genetics , ADP-Ribosylation , DNA Damage , DNA Repair , DNA/metabolism
6.
FEBS J ; 289(23): 7399-7410, 2022 12.
Article En | MEDLINE | ID: mdl-34323016

ADP-ribosylation, a modification of proteins, nucleic acids, and metabolites, confers broad functions, including roles in stress responses elicited, for example, by DNA damage and viral infection and is involved in intra- and extracellular signaling, chromatin and transcriptional regulation, protein biosynthesis, and cell death. ADP-ribosylation is catalyzed by ADP-ribosyltransferases (ARTs), which transfer ADP-ribose from NAD+ onto substrates. The modification, which occurs as mono- or poly-ADP-ribosylation, is reversible due to the action of different ADP-ribosylhydrolases. Importantly, inhibitors of ARTs are approved or are being developed for clinical use. Moreover, ADP-ribosylhydrolases are being assessed as therapeutic targets, foremost as antiviral drugs and for oncological indications. Due to the development of novel reagents and major technological advances that allow the study of ADP-ribosylation in unprecedented detail, an increasing number of cellular processes and pathways are being identified that are regulated by ADP-ribosylation. In addition, characterization of biochemical and structural aspects of the ARTs and their catalytic activities have expanded our understanding of this protein family. This increased knowledge requires that a common nomenclature be used to describe the relevant enzymes. Therefore, in this viewpoint, we propose an updated and broadly supported nomenclature for mammalian ARTs that will facilitate future discussions when addressing the biochemistry and biology of ADP-ribosylation. This is combined with a brief description of the main functions of mammalian ARTs to illustrate the increasing diversity of mono- and poly-ADP-ribose mediated cellular processes.


ADP Ribose Transferases , Protein Biosynthesis , ADP Ribose Transferases/genetics , Adenosine Diphosphate Ribose , Adenosine Diphosphate
7.
Nucleic Acids Res ; 49(18): 10477-10492, 2021 10 11.
Article En | MEDLINE | ID: mdl-34508355

ADP-ribosylation is a modification that targets a variety of macromolecules and regulates a diverse array of important cellular processes. ADP-ribosylation is catalysed by ADP-ribosyltransferases and reversed by ADP-ribosylhydrolases. Recently, an ADP-ribosyltransferase toxin termed 'DarT' from bacteria, which is distantly related to human PARPs, was shown to modify thymidine in single-stranded DNA in a sequence specific manner. The antitoxin of DarT is the macrodomain containing ADP-ribosylhydrolase DarG, which shares striking structural homology with the human ADP-ribosylhydrolase TARG1. Here, we show that TARG1, like DarG, can reverse thymidine-linked DNA ADP-ribosylation. We find that TARG1-deficient human cells are extremely sensitive to DNA ADP-ribosylation. Furthermore, we also demonstrate the first detection of reversible ADP-ribosylation on genomic DNA in vivo from human cells. Collectively, our results elucidate the impact of DNA ADP-ribosylation in human cells and provides a molecular toolkit for future studies into this largely unknown facet of ADP-ribosylation.


Adenosine Diphosphate Ribose/metabolism , DNA/metabolism , Thiolester Hydrolases/physiology , Adenosine Diphosphate Ribose/chemistry , Bacterial Toxins/metabolism , Cell Line , DNA Repair , DNA Replication , DNA, Single-Stranded/metabolism , Humans , Thiolester Hydrolases/chemistry , Thymidine/metabolism
8.
Sci Adv ; 6(51)2020 12.
Article En | MEDLINE | ID: mdl-33355125

Poly(ADP-ribose) polymerase (PARP) inhibitors are used in the treatment of BRCA-deficient cancers, with treatments currently extending toward other homologous recombination defective tumors. In a genome-wide CRISPR knockout screen with olaparib, we identify ALC1 (Amplified in Liver Cancer 1)-a cancer-relevant poly(ADP-ribose)-regulated chromatin remodeling enzyme-as a key modulator of sensitivity to PARP inhibitor. We found that ALC1 can remove inactive PARP1 indirectly through binding to PARylated chromatin. Consequently, ALC1 deficiency enhances trapping of inhibited PARP1, which then impairs the binding of both nonhomologous end-joining and homologous recombination repair factors to DNA lesions. We also establish that ALC1 overexpression, a common feature in multiple tumor types, reduces the sensitivity of BRCA-deficient cells to PARP inhibitors. Together, we conclude that ALC1-dependent PARP1 mobilization is a key step underlying PARP inhibitor resistance.


Chromatin , DNA Helicases , DNA-Binding Proteins , Poly(ADP-ribose) Polymerase Inhibitors , Cell Line, Tumor , Chromatin/genetics , Chromatin Assembly and Disassembly , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism
9.
Nucleic Acids Res ; 47(21): 11250-11267, 2019 12 02.
Article En | MEDLINE | ID: mdl-31566235

The addition of poly(ADP-ribose) (PAR) chains along the chromatin fiber due to PARP1 activity regulates the recruitment of multiple factors to sites of DNA damage. In this manuscript, we investigated how, besides direct binding to PAR, early chromatin unfolding events controlled by PAR signaling contribute to recruitment to DNA lesions. We observed that different DNA-binding, but not histone-binding, domains accumulate at damaged chromatin in a PAR-dependent manner, and that this recruitment correlates with their affinity for DNA. Our findings indicate that this recruitment is promoted by early PAR-dependent chromatin remodeling rather than direct interaction with PAR. Moreover, recruitment is not the consequence of reduced molecular crowding at unfolded damaged chromatin but instead originates from facilitated binding to more exposed DNA. These findings are further substantiated by the observation that PAR-dependent chromatin remodeling at DNA lesions underlies increased DNAse hypersensitivity. Finally, the relevance of this new mode of PAR-dependent recruitment to DNA lesions is demonstrated by the observation that reducing the affinity for DNA of both CHD4 and HP1α, two proteins shown to be involved in the DNA-damage response, strongly impairs their recruitment to DNA lesions.


Chromatin Assembly and Disassembly/physiology , Chromatin/metabolism , DNA Damage/genetics , DNA-Binding Proteins/metabolism , DNA/metabolism , Poly Adenosine Diphosphate Ribose/physiology , Binding Sites/genetics , Cells, Cultured , Chromatin/chemistry , Chromobox Protein Homolog 5 , Humans , Nucleic Acid Conformation , Poly Adenosine Diphosphate Ribose/metabolism , Protein Binding
10.
Cell Death Dis ; 10(4): 302, 2019 04 03.
Article En | MEDLINE | ID: mdl-30944311

Severe side effects often restrict clinical application of the widely used chemotherapeutic drug doxorubicin. In order to decrease required substance concentrations, new concepts for successful combination therapy are needed. Since doxorubicin causes DNA damage, combination with compounds that modulate DNA repair could be a promising strategy. Very recently, a role of nuclear actin for DNA damage repair has been proposed, making actin a potential target for cancer therapy in combination with DNA-damaging therapeutics. This is of special interest, since actin-binding compounds have not yet found their way into clinics. We find that low-dose combination treatment of doxorubicin with the actin polymerizer chondramide B (ChB) synergistically inhibits tumor growth in vivo. On the cellular level we demonstrate that actin binders inhibit distinctive double strand break (DSB) repair pathways. Actin manipulation impairs the recruitment of replication factor A (RPA) to the site of damage, a process crucial for homologous recombination. In addition, actin binders reduce autophosphorylation of DNA-dependent protein kinase (DNA-PK) during nonhomologous end joining. Our findings substantiate a direct involvement of actin in nuclear DSB repair pathways, and propose actin as a therapeutic target for combination therapy with DNA-damaging agents such as doxorubicin.


Actins/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bacterial Proteins/therapeutic use , DNA Damage/drug effects , DNA Repair/drug effects , Depsipeptides/therapeutic use , Doxorubicin/therapeutic use , Actins/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bacterial Proteins/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cell Death/drug effects , DNA End-Joining Repair/drug effects , DNA-Activated Protein Kinase/metabolism , Depsipeptides/pharmacology , Doxorubicin/pharmacology , HeLa Cells , Humans , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Recombination, Genetic/drug effects , Replication Protein A/genetics , Replication Protein A/metabolism , Thiazolidines/pharmacology , Thiazolidines/therapeutic use , Transplantation, Heterologous
11.
EMBO Rep ; 19(10)2018 10.
Article En | MEDLINE | ID: mdl-30177554

MacroH2A histone variants suppress tumor progression and act as epigenetic barriers to induced pluripotency. How they impart their influence on chromatin plasticity is not well understood. Here, we analyze how the different domains of macroH2A proteins contribute to chromatin structure and dynamics. By solving the crystal structure of the macrodomain of human macroH2A2 at 1.7 Å, we find that its putative binding pocket exhibits marked structural differences compared with the macroH2A1.1 isoform, rendering macroH2A2 unable to bind ADP-ribose. Quantitative binding assays show that this specificity is conserved among vertebrate macroH2A isoforms. We further find that macroH2A histones reduce the transient, PARP1-dependent chromatin relaxation that occurs in living cells upon DNA damage through two distinct mechanisms. First, macroH2A1.1 mediates an isoform-specific effect through its ability to suppress PARP1 activity. Second, the unstructured linker region exerts an additional repressive effect that is common to all macroH2A proteins. In the absence of DNA damage, the macroH2A linker is also sufficient for rescuing heterochromatin architecture in cells deficient for macroH2A.


Chromatin/genetics , Epigenesis, Genetic/genetics , Histones/chemistry , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/genetics , Chromatin/chemistry , Crystallography, X-Ray , DNA Damage/genetics , Heterochromatin/chemistry , Heterochromatin/genetics , Histones/genetics , Humans , Poly (ADP-Ribose) Polymerase-1/chemistry , Poly (ADP-Ribose) Polymerase-1/genetics , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/genetics
12.
Methods Mol Biol ; 1813: 11-24, 2018.
Article En | MEDLINE | ID: mdl-30097858

Poly(ADP-ribosyl)ation (PARylation) is a dynamic posttranslational modification that is added and removed rapidly at sites of DNA damage. PARylation is important for numerous aspects of DNA repair including chromatin decondensation and protein recruitment. Visualization of PARylation levels after DNA damage induction is generally obtained using traditional immunofluorescent techniques on fixed cells, which results in limited temporal resolution. Here, we describe a microscopy-based method to track ADP-ribosylation at break sites. This method relies on DNA damage induction using a 405 nm FRAP laser on Hoechst-treated cells expressing GFP-tagged PAR-binding proteins, such as macrodomains where the recruitment of the PAR-binder to sites of DNA damage gives an indication of PARylation levels.


Adenosine Diphosphate Ribose/isolation & purification , Microscopy/methods , Molecular Biology/methods , Adenosine Diphosphate Ribose/genetics , DNA Damage/genetics , DNA Repair/genetics , Poly ADP Ribosylation/genetics , Protein Binding/genetics , Protein Processing, Post-Translational/genetics
13.
Nucleic Acids Res ; 46(12): 6087-6098, 2018 07 06.
Article En | MEDLINE | ID: mdl-29733391

One of the first events to occur upon DNA damage is the local opening of the compact chromatin architecture, facilitating access of repair proteins to DNA lesions. This early relaxation is triggered by poly(ADP-ribosyl)ation by PARP1 in addition to ATP-dependent chromatin remodeling. CHD4 recruits to DNA breaks in a PAR-dependent manner, although it lacks any recognizable PAR-binding domain, and has the ability to relax chromatin structure. However, its role in chromatin relaxation at the site of DNA damage has not been explored. Using a live cell fluorescence three-hybrid assay, we demonstrate that the recruitment of CHD4 to DNA damage, while being poly(ADP-ribosyl)ation-dependent, is not through binding poly(ADP-ribose). Additionally, we show that CHD3 is recruited to DNA breaks in the same manner as CHD4 and that both CHD3 and CHD4 play active roles in chromatin remodeling at DNA breaks. Together, our findings reveal a two-step mechanism for DNA damage induced chromatin relaxation in which PARP1 and the PAR-binding remodeler activities of Alc1/CHD1L induce an initial chromatin relaxation phase that promotes the subsequent recruitment of CHD3 and CHD4 via binding to DNA for further chromatin remodeling at DNA breaks.


Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA Breaks , DNA Helicases/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Cell Line , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/physiology , Signal Transduction
14.
Mol Cell ; 68(5): 860-871.e7, 2017 Dec 07.
Article En | MEDLINE | ID: mdl-29220653

DNA damage triggers chromatin remodeling by mechanisms that are poorly understood. The oncogene and chromatin remodeler ALC1/CHD1L massively decompacts chromatin in vivo yet is inactive prior to DNA-damage-mediated PARP1 induction. We show that the interaction of the ALC1 macrodomain with the ATPase module mediates auto-inhibition. PARP1 activation suppresses this inhibitory interaction. Crucially, release from auto-inhibition requires a poly-ADP-ribose (PAR) binding macrodomain. We identify tri-ADP-ribose as a potent PAR-mimic and synthetic allosteric effector that abrogates ATPase-macrodomain interactions, promotes an ungated conformation, and activates the remodeler's ATPase. ALC1 fragments lacking the regulatory macrodomain relax chromatin in vivo without requiring PARP1 activation. Further, the ATPase restricts the macrodomain's interaction with PARP1 under non-DNA damage conditions. Somatic cancer mutants disrupt ALC1's auto-inhibition and activate chromatin remodeling. Our data show that the NAD+-metabolite and nucleic acid PAR triggers ALC1 to drive chromatin relaxation. Modular allostery in this oncogene tightly controls its robust, DNA-damage-dependent activation.


Chromatin Assembly and Disassembly , DNA Damage , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Neoplasms/enzymology , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Allosteric Regulation , Binding Sites , Cell Line, Tumor , DNA Helicases/chemistry , DNA Helicases/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Enzyme Activation , Humans , Mutation , Neoplasms/genetics , Neoplasms/pathology , Nucleic Acid Conformation , Poly (ADP-Ribose) Polymerase-1/chemistry , Poly (ADP-Ribose) Polymerase-1/genetics , Poly ADP Ribosylation , Poly Adenosine Diphosphate Ribose/chemistry , Protein Binding , Structure-Activity Relationship , Time Factors
15.
Elife ; 62017 08 10.
Article En | MEDLINE | ID: mdl-28796599

The modification of serines by molecules of ADP-ribose plays an important role in signaling that the DNA in a cell has been damaged and needs to be repaired.


ADP-Ribosylation , Serine , Adenosine Diphosphate Ribose , Hydrolases , Signal Transduction
16.
Methods Mol Biol ; 1608: 165-183, 2017.
Article En | MEDLINE | ID: mdl-28695510

The tightly packed and dynamic structure of chromatin can undergo major reorganization in response to endogenous or exogenous stimuli, such as the regulation of transcription or the cell cycle, or following DNA damage. A fast and local chromatin decondensation is observed upon DNA damage induced by laser micro-irradiation. This decondensation is under the control of poly(ADP-ribosyl)ation (PARylation) by PARP1, one of the first proteins recruited at the DNA damage sites. This chapter provides a step-by-step guide to perform and analyze chromatin decondensation upon DNA damage induction. The protocol is based on fluorescence microscopy of live cells expressing a core histone tagged with a photoactivatable fluorophore. Laser micro-irradiation is used to simultaneously induce DNA damage and activate the fluorescence signal within the irradiated area. This photo-perturbation experiment can be easily implemented on any confocal laser-scanning microscope equipped with a photoperturbation module. The experimental framework can also be used to follow chromatin relaxation in parallel with the recruitment kinetics of a protein of interest at DNA lesions in cells co-expressing the tagged histones and a second protein of interest fused to a different fluorescent tag.


Chromatin Assembly and Disassembly/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Animals , DNA Damage/genetics , DNA Repair/genetics , DNA Repair/physiology , Humans , Poly (ADP-Ribose) Polymerase-1/genetics , Poly ADP Ribosylation/genetics , Poly ADP Ribosylation/physiology
17.
Nucleic Acids Res ; 45(1): 244-254, 2017 Jan 09.
Article En | MEDLINE | ID: mdl-28069995

ADP-ribosylation is a dynamic post-translation modification that regulates the early phase of various DNA repair pathways by recruiting repair factors to chromatin. ADP-ribosylation levels are defined by the activities of specific transferases and hydrolases. However, except for the transferase PARP1/ARDT1 little is known about regulation of these enzymes. We found that MacroD2, a mono-ADP-ribosylhydrolase, is exported from the nucleus upon DNA damage, and that this nuclear export is induced by ATM activity. We show that the export is dependent on the phosphorylation of two SQ/TQ motifs, suggesting a novel direct interaction between ATM and ADP-ribosylation. Lastly, we show that MacroD2 nuclear export temporally restricts its recruitment to DNA lesions, which may decrease the net ADP-ribosylhydrolase activity at the site of DNA damage. Together, our results identify a novel feedback regulation between two crucial DNA damage-induced signaling pathways: ADP-ribosylation and ATM activation.


Adenosine Diphosphate/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , DNA Damage , DNA Repair Enzymes/genetics , Hydrolases/genetics , Poly(ADP-ribose) Polymerases/genetics , Protein Processing, Post-Translational , Active Transport, Cell Nucleus , Amino Acid Motifs , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , DNA Repair Enzymes/metabolism , Feedback, Physiological , HeLa Cells , Humans , Hydrolases/metabolism , Osteoblasts , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction
18.
Mol Biol Cell ; 27(24): 3791-3799, 2016 12 01.
Article En | MEDLINE | ID: mdl-27733626

Chromatin relaxation is one of the earliest cellular responses to DNA damage. However, what determines these structural changes, including their ATP requirement, is not well understood. Using live-cell imaging and laser microirradiation to induce DNA lesions, we show that the local chromatin relaxation at DNA damage sites is regulated by PARP1 enzymatic activity. We also report that H1 is mobilized at DNA damage sites, but, since this mobilization is largely independent of poly(ADP-ribosyl)ation, it cannot solely explain the chromatin relaxation. Finally, we demonstrate the involvement of Alc1, a poly(ADP-ribose)- and ATP-dependent remodeler, in the chromatin-relaxation process. Deletion of Alc1 impairs chromatin relaxation after DNA damage, while its overexpression strongly enhances relaxation. Altogether our results identify Alc1 as an important player in the fast kinetics of the NAD+- and ATP-dependent chromatin relaxation upon DNA damage in vivo.


DNA Helicases/metabolism , DNA Helicases/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Poly Adenosine Diphosphate Ribose/metabolism , Cell Culture Techniques , Chromatin/physiology , Chromatin Assembly and Disassembly/physiology , DNA , DNA Damage , DNA Repair/physiology , Histones/metabolism , Humans , Nucleosomes , Optical Imaging , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly Adenosine Diphosphate Ribose/physiology , Poly(ADP-ribose) Polymerases/metabolism
19.
Nucleic Acids Res ; 44(7): 3105-17, 2016 Apr 20.
Article En | MEDLINE | ID: mdl-26673727

Eukaryotic chromatin is a complex yet dynamic structure, which is regulated in part by the assembly and disassembly of nucleosomes. Key to this process is a group of proteins termed histone chaperones that guide the thermodynamic assembly of nucleosomes by interacting with soluble histones. Here we investigate the interaction between the histone chaperone sNASP and its histone H3 substrate. We find that sNASP binds with nanomolar affinity to a conserved heptapeptide motif in the globular domain of H3, close to the C-terminus. Through functional analysis of sNASP homologues we identified point mutations in surface residues within the TPR domain of sNASP that disrupt H3 peptide interaction, but do not completely disrupt binding to full length H3 in cells, suggesting that sNASP interacts with H3 through additional contacts. Furthermore, chemical shift perturbations from(1)H-(15)N HSQC experiments show that H3 peptide binding maps to the helical groove formed by the stacked TPR motifs of sNASP. Our findings reveal a new mode of interaction between a TPR repeat domain and an evolutionarily conserved peptide motif found in canonical H3 and in all histone H3 variants, including CenpA and have implications for the mechanism of histone chaperoning within the cell.


Autoantigens/chemistry , Autoantigens/metabolism , Histone Chaperones/chemistry , Histone Chaperones/metabolism , Histones/chemistry , Histones/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Conserved Sequence , Peptides/chemistry , Protein Binding , Protein Structure, Secondary , Repetitive Sequences, Amino Acid
...