Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Mol Cell ; 48(5): 785-98, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23102699

ABSTRACT

Poly-(ADP-ribose) glycohydrolase (PARG) is a catabolic enzyme that cleaves ADP-ribose polymers synthesized by poly-(ADP-ribose) polymerases. Here, transcriptome profiling and differentiation assay revealed a requirement of PARG for retinoic acid receptor (RAR)-mediated transcription. Mechanistically, PARG accumulates early at promoters of RAR-responsive genes upon retinoic acid treatment to promote the formation of an appropriate chromatin environment suitable for transcription. Silencing of PARG or knockout of its enzymatic activity maintains the H3K9me2 mark at the promoter of the RAR-dependent genes, leading to the absence of preinitiation complex formation. In the absence of PARG, we found that the H3K9 demethylase KDM4D/JMJD2D became PARsylated. Mutation of two glutamic acids located in the Jumonji N domain of KDM4D inhibited PARsylation. PARG becomes dispensable for ligand-dependent transcription when either a PARP inhibitor or a non-PARsylable KDM4D/JMJD2D mutant is used. Our results define PARG as a coactivator regulating chromatin remodeling during RA-dependent gene expression.


Subject(s)
Glycoside Hydrolases/metabolism , Receptors, Retinoic Acid/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Chromatin Assembly and Disassembly , Enzyme Inhibitors/pharmacology , Gene Expression Profiling/methods , Gene Expression Regulation , Glutamic Acid , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/genetics , HeLa Cells , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Methylation , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , RNA Interference , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/metabolism , Retinoic Acid Receptor alpha , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection , Tretinoin/pharmacology
2.
Eur J Immunol ; 47(4): 665-676, 2017 04.
Article in English | MEDLINE | ID: mdl-28105679

ABSTRACT

To mount highly specific and adapted immune responses, B lymphocytes assemble and diversify their antibody repertoire through mechanisms involving the formation of programmed DNA damage. Immunoglobulin class switch recombination (CSR) is triggered by DNA lesions induced by activation-induced cytidine deaminase, which are processed to double-stranded DNA break (DSB) intermediates. These DSBs activate the cellular DNA damage response and enroll numerous DNA repair factors, involving poly(ADP-ribose) polymerases Parp1, Parp2, and Parp3 to promote appropriate DNA repair and efficient long-range recombination. The macroParp Parp9, which is overexpressed in certain lymphomas, has been recently implicated in DSB repair, acting together with Parp1. Here, we examine the contribution of Parp9 to the resolution of physiological DSBs incurred during V(D)J recombination and CSR by generating Parp9-/- mice. We find that Parp9-deficient mice are viable, fertile, and do not show any overt phenotype. Moreover, we find that Parp9 is dispensable for B-cell development. Finally, we show that CSR and DNA end-joining are robust in the absence of Parp9, indicating that Parp9 is not essential in vivo to achieve physiological DSB repair, or that strong compensatory mechanisms exist.


Subject(s)
B-Lymphocytes/physiology , DNA End-Joining Repair , Immunoglobulin Class Switching , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Adaptive Immunity , Animals , Cells, Cultured , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair , Immunoglobulins/genetics , Immunoglobulins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics
3.
J Biol Chem ; 291(10): 4873-81, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26772194

ABSTRACT

The epigenetic information encoded in the genomic DNA methylation pattern is translated by methylcytosine binding proteins like MeCP2 into chromatin topology and structure and gene activity states. We have shown previously that the MeCP2 level increases during differentiation and that it causes large-scale chromatin reorganization, which is disturbed by MeCP2 Rett syndrome mutations. Phosphorylation and other posttranslational modifications of MeCP2 have been described recently to modulate its function. Here we show poly(ADP-ribosyl)ation of endogenous MeCP2 in mouse brain tissue. Consequently, we found that MeCP2 induced aggregation of pericentric heterochromatin and that its chromatin accumulation was enhanced in poly(ADP-ribose) polymerase (PARP) 1(-/-) compared with wild-type cells. We mapped the poly(ADP-ribosyl)ation domains and engineered MeCP2 mutation constructs to further analyze potential effects on DNA binding affinity and large-scale chromatin remodeling. Single or double deletion of the poly(ADP-ribosyl)ated regions and PARP inhibition increased the heterochromatin clustering ability of MeCP2. Increased chromatin clustering may reflect increased binding affinity. In agreement with this hypothesis, we found that PARP-1 deficiency significantly increased the chromatin binding affinity of MeCP2 in vivo. These data provide novel mechanistic insights into the regulation of MeCP2-mediated, higher-order chromatin architecture and suggest therapeutic opportunities to manipulate MeCP2 function.


Subject(s)
Chromatin/chemistry , Methyl-CpG-Binding Protein 2/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational , Animals , Binding Sites , Brain/metabolism , Chromatin/metabolism , HEK293 Cells , Humans , Methyl-CpG-Binding Protein 2/chemistry , Methyl-CpG-Binding Protein 2/genetics , Mice , Mutation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Binding , Rats , Sf9 Cells , Spodoptera
4.
Mol Cell ; 36(3): 365-78, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19917246

ABSTRACT

Upon genotoxic stresses, cells activate IkappaB kinases (IKKs) and the transcription factor NF-kappaB to modulate apoptotic responses. The SUMO-1 ligase PIASy and the kinase ataxia talengiectasia mutated (ATM) have been implicated to SUMOylate and phosphorylate nuclear IKKgamma (NEMO) in a consecutive mode of action, which in turn results in activation of cytoplasmic IKK holocomplexes. However, the nuclear signals and scaffold structures that initiate IKKgamma recruitment and activation are unknown. Here, we show that poly(ADP-ribose)-polymerase-1 (PARP-1) is the DNA proximal regulator, which senses DNA strand breaks and, through poly(ADP-ribose) (PAR) synthesis, assembles IKKgamma, PIASy, and ATM in a dynamic manner. Signalosome formation involves direct protein-protein interactions and binding to ADP-ribose polymers through PAR binding motifs (PARBM). Activated PARP-1 and a PARBM in PIASy are required to trigger IKKgamma SUMOylation, which in turn permits IKK and NF-kappaB activation, as well as NF-kappaB-regulated resistance to apoptosis.


Subject(s)
DNA Damage , I-kappa B Kinase/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , I-kappa B Kinase/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Molecular Sequence Data , NF-kappa B/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Protein Serine-Threonine Kinases/metabolism , Sequence Homology, Amino Acid , Tumor Suppressor Proteins/metabolism
5.
Trends Biochem Sci ; 37(9): 381-90, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22766145

ABSTRACT

Poly(ADP-ribosyl)ation (PARylation), a protein post-translational modification that was originally connected to the DNA damage response, is now known to engage in a continuously increasing number of biological processes. Despite extensive research and ceaseless, important findings about its role and mode of action, poly(ADP-ribose) remains an enigma regarding its structural complexity and diversity. The recent identification and structural characterization of four different poly(ADP-ribose) binding motifs represents a quantum leap in the comprehension of how this molecule can be decoded. Moreover, the recent discovery of a direct connection between PARylation and poly-ubiquitylation in targeting proteins for degradation by the proteasome has paved the way for a new interpretation of this protein modification. These two novel aspects, poly(ADP-ribose) recognition and readout by the ubiquitylation/proteasome system are developed here.


Subject(s)
Poly Adenosine Diphosphate Ribose/metabolism , Animals , DNA Damage , Humans , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Ubiquitination
6.
Cancer Cell Int ; 16: 53, 2016.
Article in English | MEDLINE | ID: mdl-27375368

ABSTRACT

BACKGROUND: Poly(ADP-ribose) polymerase (PARP) inhibitors have entered the clinics for their promising anticancer effect as adjuvant in chemo- and radiotherapy and as single agent on BRCA-mutated tumours. Poly(ADP-ribose) glycohydrolase (PARG) deficiency was also shown to potentiate the cytotoxicity of genotoxic agents and irradiation. The aim of this study is to investigate the effect of PARG deficiency on BRCA1- and/or PTEN-deficient tumour cells. METHODS: Since no PARG inhibitors are available for in vivo studies, PARG was depleted by siRNA in several cancer cell lines, proficient or deficient for BRCA1 and/or PTEN. The impact on cell survival was evaluated by colony formation assay and short-term viability assays. The effect of simultaneous PARG and BRCA1 depletion on homologous recombination (HR) efficacy was evaluated by immunodetection of RAD51 foci and using an in vivo HR assay. RESULTS: The BRCA1-deficient cell lines MDA-MB-436, HCC1937 and UWB1.289 showed mild sensitivity to PARG depletion, whereas no sensitivity was observed for the BRCA1-proficient MDA-MB-231, MDA-MB-468, MCF10A and U2OS cell lines. However, the BRCA1-reconstituted UWB1.289 cell lines was similarly sensitive to PARG depletion than the BRCA1-deficient UWB1.289, and the simultaneous depletion of PARG and BRCA1 and/or PTEN in MDA-MB-231 or U2OS cells was not more cytotoxic than depletion of BRCA1 or PTEN only. CONCLUSIONS: Some tumour cells displayed slight sensitivity to PARG deficiency, but this sensitivity could not be correlated to BRCA1- or PTEN-deficiency. Therefore, PARG depletion cannot be considered as a strategy to kill tumours cells mutated in BRCA1 or PTEN.

7.
Nucleic Acids Res ; 42(12): 7776-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24906880

ABSTRACT

Poly(ADP-ribosyl)ation is involved in numerous bio-logical processes including DNA repair, transcription and cell death. Cellular levels of poly(ADP-ribose) (PAR) are regulated by PAR polymerases (PARPs) and the degrading enzyme PAR glycohydrolase (PARG), controlling the cell fate decision between life and death in response to DNA damage. Replication stress is a source of DNA damage, leading to transient stalling of replication forks or to their collapse followed by the generation of double-strand breaks (DSB). The involvement of PARP-1 in replicative stress response has been described, whereas the consequences of a deregulated PAR catabolism are not yet well established. Here, we show that PARG-deprived cells showed an enhanced sensitivity to the replication inhibitor hydroxyurea. PARG is dispensable to recover from transient replicative stress but is necessary to avoid massive PAR production upon prolonged replicative stress, conditions leading to fork collapse and DSB. Extensive PAR accumulation impairs replication protein A association with collapsed forks resulting in compromised DSB repair via homologous recombination. Our results highlight the critical role of PARG in tightly controlling PAR levels produced upon genotoxic stress to prevent the detrimental effects of PAR over-accumulation.


Subject(s)
DNA Repair , DNA Replication , Glycoside Hydrolases/physiology , Poly Adenosine Diphosphate Ribose/metabolism , Cell Line , Chromatin/metabolism , DNA, Single-Stranded/analysis , HeLa Cells , Histones/metabolism , Humans , Hydroxyurea/pharmacology , Phosphorylation , Poly(ADP-ribose) Polymerase Inhibitors , Recombinational DNA Repair , Replication Protein A/metabolism , S Phase/drug effects , S Phase Cell Cycle Checkpoints , Stress, Physiological/genetics
8.
Nucleic Acids Res ; 42(7): 4435-49, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24493735

ABSTRACT

Poly(ADP-ribose) polymerases (PARP) attach poly(ADP-ribose) (PAR) chains to various proteins including themselves and chromatin. Topoisomerase I (Top1) regulates DNA supercoiling and is the target of camptothecin and indenoisoquinoline anticancer drugs, as it forms Top1 cleavage complexes (Top1cc) that are trapped by the drugs. Endogenous and carcinogenic DNA lesions can also trap Top1cc. Tyrosyl-DNA phosphodiesterase 1 (TDP1), a key repair enzyme for trapped Top1cc, hydrolyzes the phosphodiester bond between the DNA 3'-end and the Top1 tyrosyl moiety. Alternative repair pathways for Top1cc involve endonuclease cleavage. However, it is unknown what determines the choice between TDP1 and the endonuclease repair pathways. Here we show that PARP1 plays a critical role in this process. By generating TDP1 and PARP1 double-knockout lymphoma chicken DT40 cells, we demonstrate that TDP1 and PARP1 are epistatic for the repair of Top1cc. The N-terminal domain of TDP1 directly binds the C-terminal domain of PARP1, and TDP1 is PARylated by PARP1. PARylation stabilizes TDP1 together with SUMOylation of TDP1. TDP1 PARylation enhances its recruitment to DNA damage sites without interfering with TDP1 catalytic activity. TDP1-PARP1 complexes, in turn recruit X-ray repair cross-complementing protein 1 (XRCC1). This work identifies PARP1 as a key component driving the repair of trapped Top1cc by TDP1.


Subject(s)
DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Phosphoric Diester Hydrolases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , Epistasis, Genetic , Humans , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Protein Interaction Domains and Motifs , Sumoylation , X-ray Repair Cross Complementing Protein 1
9.
Nucleic Acids Res ; 42(9): 5616-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24598253

ABSTRACT

The repair of toxic double-strand breaks (DSB) is critical for the maintenance of genome integrity. The major mechanisms that cope with DSB are: homologous recombination (HR) and classical or alternative nonhomologous end joining (C-NHEJ versus A-EJ). Because these pathways compete for the repair of DSB, the choice of the appropriate repair pathway is pivotal. Among the mechanisms that influence this choice, deoxyribonucleic acid (DNA) end resection plays a critical role by driving cells to HR, while accurate C-NHEJ is suppressed. Furthermore, end resection promotes error-prone A-EJ. Increasing evidence define Poly(ADP-ribose) polymerase 3 (PARP3, also known as ARTD3) as an important player in cellular response to DSB. In this work, we reveal a specific feature of PARP3 that together with Ku80 limits DNA end resection and thereby helps in making the choice between HR and NHEJ pathways. PARP3 interacts with and PARylates Ku70/Ku80. The depletion of PARP3 impairs the recruitment of YFP-Ku80 to laser-induced DNA damage sites and induces an imbalance between BRCA1 and 53BP1. Both events result in compromised accurate C-NHEJ and a concomitant increase in DNA end resection. Nevertheless, HR is significantly reduced upon PARP3 silencing while the enhanced end resection causes mutagenic deletions during A-EJ. As a result, the absence of PARP3 confers hypersensitivity to anti-tumoral drugs generating DSB.


Subject(s)
Cell Cycle Proteins/physiology , DNA End-Joining Repair , Poly(ADP-ribose) Polymerases/physiology , Recombinational DNA Repair , Antigens, Nuclear/metabolism , Antineoplastic Agents/pharmacology , BRCA1 Protein/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Drug Screening Assays, Antitumor , Etoposide/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ku Autoantigen , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , Protein Transport , Replication Protein A/metabolism , Tumor Suppressor p53-Binding Protein 1
10.
J Biol Chem ; 289(23): 16223-38, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24782312

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP1, also known as ARTD1) is an abundant nuclear enzyme that plays important roles in DNA repair, gene transcription, and differentiation through the modulation of chromatin structure and function. In this work we identify a physical and functional poly(ADP-ribose)-mediated interaction of PARP1 with the E3 ubiquitin ligase UHRF1 (also known as NP95, ICBP90) that influences two UHRF1-regulated cellular processes. On the one hand, we uncovered a cooperative interplay between PARP1 and UHRF1 in the accumulation of the heterochromatin repressive mark H4K20me3. The absence of PARP1 led to reduced accumulation of H4K20me3 onto pericentric heterochromatin that coincided with abnormally enhanced transcription. The loss of H4K20me3 was rescued by the additional depletion of UHRF1. In contrast, although PARP1 also seemed to facilitate the association of UHRF1 with DNMT1, its absence did not impair the loading of DNMT1 onto heterochromatin or the methylation of pericentric regions, possibly owing to a compensating interaction of DNMT1 with PCNA. On the other hand, we showed that PARP1 controls the UHRF1-mediated ubiquitination of DNMT1 to timely regulate its abundance during S and G2 phase. Together, this report identifies PARP1 as a novel modulator of two UHRF1-regulated heterochromatin-associated events: the accumulation of H4K20me3 and the clearance of DNMT1.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , 3T3 Cells , Animals , Base Sequence , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Primers , Mice , Protein Binding , Ubiquitin-Protein Ligases , Ubiquitination
11.
Blood ; 122(1): 44-54, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23678004

ABSTRACT

Hematopoietic stem cells self-renew for life to guarantee the continuous supply of all blood cell lineages. Here we show that Poly(ADP-ribose) polymerase-2 (Parp-2) plays an essential role in hematopoietic stem/progenitor cells (HSPC) survival under steady-state conditions and in response to stress. Increased levels of cell death were observed in HSPC from untreated Parp-2-/- mice, but this deficit was compensated by increased rates of self-renewal, associated with impaired reconstitution of hematopoiesis upon serial bone marrow transplantation. Cell death after γ-irradiation correlated with an impaired capacity to repair DNA damage in the absence of Parp-2. Upon exposure to sublethal doses of γ-irradiation, Parp-2-/- mice exhibited bone marrow failure that correlated with reduced long-term repopulation potential of irradiated Parp-2-/- HSPC under competitive conditions. In line with a protective role of Parp-2 against irradiation-induced apoptosis, loss of p53 or the pro-apoptotic BH3-only protein Puma restored survival of irradiated Parp-2-/- mice, whereas loss of Noxa had no such effect. Our results show that Parp-2 plays essential roles in the surveillance of genome integrity of HSPC by orchestrating DNA repair and restraining p53-induced and Puma-mediated apoptosis. The data may affect the design of drugs targeting Parp proteins and the improvement of radiotherapy-based therapeutic strategies.


Subject(s)
Gamma Rays/adverse effects , Hematopoiesis/physiology , Hematopoiesis/radiation effects , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/physiology , Anemia, Aplastic , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/physiology , Bone Marrow Diseases , Bone Marrow Failure Disorders , Cell Survival/physiology , Cell Survival/radiation effects , DNA Damage/physiology , DNA Repair/physiology , Female , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/physiopathology , Homeostasis/physiology , Homeostasis/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/physiopathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
12.
Exp Cell Res ; 329(1): 18-25, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25017100

ABSTRACT

Poly(ADP-ribosyl)ation (PARylation) is a post-translational modification of proteins catalysed by Poly(ADP-ribose) polymerases (PARP). A wealth of recent advances in the biochemical and functional characterization of the DNA-dependent PARP family members have highlighted their key contribution in the DNA damage response network, the best characterized being the role of PARP1 and PARP2 in the resolution of single-strand breaks as part of the BER/SSBR process. How PARylation contributes to the repair of double-strand breaks is less well defined but has become recently the subject of significant research in the field. The aim of this review is to provide an overview of the current knowledge concerning the role of the DNA-activated PARP1, PARP2 and PARP3 in cellular response to double-strand breaks (DSB). In addition, we outline the biological significance of these properties in response to programmed DNA lesions formed during physiological processes such as antibody repertoire assembly and diversification.


Subject(s)
DNA Damage/genetics , DNA Repair , DNA/genetics , Poly(ADP-ribose) Polymerases/metabolism , Animals , Humans
13.
Proc Natl Acad Sci U S A ; 108(7): 2783-8, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21270334

ABSTRACT

The ADP ribosyl transferase [poly(ADP-ribose) polymerase] ARTD3(PARP3) is a newly characterized member of the ARTD(PARP) family that catalyzes the reaction of ADP ribosylation, a key posttranslational modification of proteins involved in different signaling pathways from DNA damage to energy metabolism and organismal memory. This enzyme shares high structural similarities with the DNA repair enzymes PARP1 and PARP2 and accordingly has been found to catalyse poly(ADP ribose) synthesis. However, relatively little is known about its in vivo cellular properties. By combining biochemical studies with the generation and characterization of loss-of-function human and mouse models, we describe PARP3 as a newcomer in genome integrity and mitotic progression. We report a particular role of PARP3 in cellular response to double-strand breaks, most likely in concert with PARP1. We identify PARP3 as a critical player in the stabilization of the mitotic spindle and in telomere integrity notably by associating and regulating the mitotic components NuMA and tankyrase 1. Both functions open stimulating prospects for specifically targeting PARP3 in cancer therapy.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , Genomic Instability/genetics , Mitosis/genetics , Poly(ADP-ribose) Polymerases/metabolism , Spindle Apparatus/physiology , Adenosine Diphosphate/metabolism , Animals , Antigens, Nuclear/metabolism , Blotting, Western , Cell Line, Tumor , Colony-Forming Units Assay , Comet Assay , DNA Primers/genetics , Fluorescent Antibody Technique, Indirect , Genomic Instability/physiology , Humans , Immunoprecipitation , In Situ Hybridization, Fluorescence , Mass Spectrometry , Mice , Mice, Knockout , Microscopy, Video , Mitosis/physiology , Nuclear Matrix-Associated Proteins/metabolism , Poly(ADP-ribose) Polymerases/deficiency , Tankyrases/metabolism
14.
EMBO J ; 28(22): 3534-48, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19779455

ABSTRACT

Heat shock and other environmental stresses rapidly induce transcriptional responses subject to regulation by a variety of post-translational modifications. Among these, poly(ADP-ribosyl)ation and sumoylation have received growing attention. Here we show that the SUMO E3 ligase PIASy interacts with the poly(ADP-ribose) polymerase PARP-1, and that PIASy mediates heat shock-induced poly-sumoylation of PARP-1. Furthermore, PIASy, and hence sumoylation, appears indispensable for full activation of the inducible HSP70.1 gene. Chromatin immunoprecipitation experiments show that PIASy, SUMO and the SUMO-conjugating enzyme Ubc9 are rapidly recruited to the HSP70.1 promoter upon heat shock, and that they are subsequently released with kinetics similar to PARP-1. Finally, we provide evidence that the SUMO-targeted ubiquitin ligase RNF4 mediates heat-shock-inducible ubiquitination of PARP-1, regulates the stability of PARP-1, and, like PIASy, is a positive regulator of HSP70.1 gene activity. These results, thus, point to a novel mechanism for regulating PARP-1 transcription function, and suggest crosstalk between sumoylation and RNF4-mediated ubiquitination in regulating gene expression in response to heat shock.


Subject(s)
Heat-Shock Response/physiology , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/physiology , SUMO-1 Protein/metabolism , Transcriptional Activation , Animals , Cells, Cultured , Gene Knockdown Techniques , HeLa Cells , Humans , Jurkat Cells , Mice , Models, Biological , Poly (ADP-Ribose) Polymerase-1 , Poly-ADP-Ribose Binding Proteins , Protein Inhibitors of Activated STAT/metabolism , Protein Inhibitors of Activated STAT/physiology , Protein Processing, Post-Translational/physiology , Spodoptera , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology
15.
Nucleic Acids Res ; 39(12): 5045-56, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21398629

ABSTRACT

Post-translational poly(ADP-ribosyl)ation has diverse essential functions in the cellular response to DNA damage as it contributes to avid DNA damage detection and assembly of the cellular repair machinery but extensive modification eventually also induces cell death. While there are 17 human poly(ADP-ribose) polymerase (PARP) genes, there is only one poly(ADP-ribose) glycohydrolase (PARG) gene encoding several PARG isoforms located in different subcellular compartments. To investigate the recruitment of PARG isoforms to DNA repair sites we locally introduced DNA damage by laser microirradiation. All PARG isoforms were recruited to DNA damage sites except for a mitochondrial localized PARG fragment. Using PARP knock out cells and PARP inhibitors, we showed that PARG recruitment was only partially dependent on PARP-1 and PAR synthesis, indicating a second, PAR-independent recruitment mechanism. We found that PARG interacts with PCNA, mapped a PCNA binding site and showed that binding to PCNA contributes to PARG recruitment to DNA damage sites. This dual recruitment mode of the only nuclear PARG via the versatile loading platform PCNA and by a PAR dependent mechanism likely contributes to the dynamic regulation of this posttranslational modification and ensures the tight control of the switch between efficient DNA repair and cell death.


Subject(s)
DNA Damage , Glycoside Hydrolases/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Animals , Biocatalysis , Cells, Cultured , Glycoside Hydrolases/analysis , Glycoside Hydrolases/chemistry , Humans , Lasers , Mice , Protein Isoforms/metabolism , Protein Structure, Tertiary
16.
Cell Rep Methods ; 3(7): 100512, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37533652

ABSTRACT

Time-specific modulation of gene expression during differentiation by transcription factors promotes cell diversity. However, estimating their dynamic regulatory activity at the single-cell level and in a high-throughput manner remains challenging. We present FateCompass, an integrative approach that utilizes single-cell transcriptomics data to identify lineage-specific transcription factors throughout differentiation. By combining a probabilistic framework with RNA velocities or differentiation potential, we estimate transition probabilities, while a linear model of gene regulation is employed to compute transcription factor activities. Considering dynamic changes and correlations of expression and activities, FateCompass identifies lineage-specific regulators. Our validation using in silico data and application to pancreatic endocrine cell differentiation datasets highlight both known and potentially novel lineage-specific regulators. Notably, we uncovered undescribed transcription factors of an enterochromaffin-like population during in vitro differentiation toward ß-like cells. FateCompass provides a valuable framework for hypothesis generation, advancing our understanding of the gene regulatory networks driving cell-fate decisions.


Subject(s)
Gene Expression Regulation , Transcription Factors , Transcription Factors/genetics , Cell Differentiation/genetics , Gene Regulatory Networks , Gene Expression Profiling
17.
J Vis Exp ; (197)2023 07 07.
Article in English | MEDLINE | ID: mdl-37486110

ABSTRACT

Genome-wide analyses with small cell populations are a major constraint for studies, particularly in the stem cell field. This work describes an efficient protocol for the fluorescence-activated cell sorting (FACS) isolation of satellite cells from the limb muscle, a tissue with a high content of structural proteins. Dissected limb muscles from adult mice were mechanically disrupted by mincing in medium supplemented with dispase and type I collagenase. Upon digestion, the homogenate was filtered through cell strainers, and cells were suspended in FACS buffer. Viability was determined with fixable viability stain, and immunostained satellite cells were isolated by FACS. Cells were lysed with Triton X-100 and released nuclei were bound to concanavalin A magnetic beads. Nucleus/bead complexes were incubated with antibodies against the transcription factor or histone modifications of interest. After washes, nucleus/bead complexes were incubated with protein A-micrococcal nuclease, and chromatin cleavage was initiated with CaCl2. After DNA extraction, libraries were generated and sequenced, and the profiles for genome-wide transcription factor binding and covalent histone modifications were obtained by bioinformatic analysis. The peaks obtained for the various histone marks showed that the binding events were specific for satellite cells. Moreover, known motif analysis unveiled that the transcription factor was bound to chromatin via its cognate response element. This protocol is therefore adapted to study gene regulation in adult mice limb muscle satellite cells.


Subject(s)
Satellite Cells, Skeletal Muscle , Mice , Animals , Flow Cytometry , Genome-Wide Association Study , Chromatin , Transcription Factors
18.
J Clin Invest ; 133(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37200107

ABSTRACT

The ADP ribosyltransferases (PARPs 1-17) regulate diverse cellular processes, including DNA damage repair. PARPs are classified on the basis of their ability to catalyze poly-ADP-ribosylation (PARylation) or mono-ADP-ribosylation (MARylation). Although PARP9 mRNA expression is significantly increased in progressive tuberculosis (TB) in humans, its participation in host immunity to TB is unknown. Here, we show that PARP9 mRNA encoding the MARylating PARP9 enzyme was upregulated during TB in humans and mice and provide evidence of a critical modulatory role for PARP9 in DNA damage, cyclic GMP-AMP synthase (cGAS) expression, and type I IFN production during TB. Thus, Parp9-deficient mice were susceptible to Mycobacterium tuberculosis infection and exhibited increased TB disease, cGAS and 2'3'-cyclic GMP-AMP (cGAMP) expression, and type I IFN production, along with upregulation of complement and coagulation pathways. Enhanced M. tuberculosis susceptibility is type I IFN dependent, as blockade of IFN α receptor (IFNAR) signaling reversed the enhanced susceptibility of Parp9-/- mice. Thus, in sharp contrast to PARP9 enhancement of type I IFN production in viral infections, this member of the MAR family plays a protective role by limiting type I IFN responses during TB.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , Humans , Mice , ADP-Ribosylation , DNA Repair , Mycobacterium tuberculosis/metabolism , Nucleotidyltransferases/genetics , Poly(ADP-ribose) Polymerases/genetics , Tuberculosis/genetics
20.
Proc Natl Acad Sci U S A ; 106(47): 19998-20003, 2009 Nov 24.
Article in English | MEDLINE | ID: mdl-19897724

ABSTRACT

Modification of eukaryotic proteins is a powerful strategy used by pathogenic bacteria to modulate host cells during infection. Previously, we demonstrated that Helicobacter pylori modify an unidentified protein within mammalian cell lysates in a manner consistent with the action of a bacterial ADP-ribosylating toxin. Here, we identified the modified eukaryotic factor as the abundant nuclear factor poly(ADP-ribose) polymerase-1 (PARP-1), which is important in the pathologies of several disease states typically associated with chronic H. pylori infection. However, rather than being ADP-ribosylated by an H. pylori toxin, the intrinsic poly(ADP-ribosyl) polymerase activity of PARP-1 is activated by a heat- and protease-sensitive H. pylori factor, resulting in automodification of PARP-1 with polymers of poly(ADP-ribose) (PAR). Moreover, during infection of gastric epithelial cells, H. pylori induce intracellular PAR-production by a PARP-1-dependent mechanism. Activation of PARP-1 by a pathogenic bacterium represents a previously unrecognized strategy for modulating host cell signaling during infection.


Subject(s)
Helicobacter pylori/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Adenosine Diphosphate Ribose/metabolism , Animals , Catalytic Domain , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gastric Mucosa/cytology , HeLa Cells , Helicobacter Infections/metabolism , Humans , Mice , Mice, Knockout , Phosphorus Radioisotopes/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL