Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Neuroinflammation ; 10: 49, 2013 Apr 22.
Article in English | MEDLINE | ID: mdl-23607899

ABSTRACT

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis characterized by entry of activated T cells and antigen presenting cells into the central nervous system and subsequent autoimmune destruction of nerve myelin. Previous studies revealed that non-selective inhibition of poly(ADP-ribose) polymerases (PARPs) 1 and 2 protect against neuroinflammation and motor dysfunction associated with EAE, but the role of the PARP-2 isoform has not yet been investigated selectively. RESULTS: EAE was induced in mice lacking PARP-2, and neurological EAE signs, blood-spine barrier (BSB) permeability, demyelination and inflammatory infiltration were monitored for 35 days after immunization. Mice lacking PARP-2 exhibited significantly reduced overall disease burden and peak neurological dysfunction. PARP-2 deletion also significantly delayed EAE onset and reduced BSB permeability, demyelination and central nervous system (CNS) markers of proinflammatory Th1 and Th17 T helper lymphocytes. CONCLUSIONS: This study represents the first description of a significant role for PARP-2 in neuroinflammation and neurological dysfunction in EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/pathology , Nervous System Diseases/pathology , Poly(ADP-ribose) Polymerases/physiology , Animals , Blood-Nerve Barrier/physiology , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/complications , Fluorescent Antibody Technique , Inflammation/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Diseases/etiology , Neutrophil Infiltration/physiology , Poly(ADP-ribose) Polymerases/genetics , T-Lymphocytes, Helper-Inducer/physiology , Th1 Cells/physiology
2.
Nucleic Acids Res ; 37(10): 3177-88, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19305001

ABSTRACT

Repair of single-stranded DNA breaks before DNA replication is critical in maintaining genomic stability; however, how cells deal with these lesions during S phase is not clear. Using combined approaches of proteomics and in vitro and in vivo protein-protein interaction, we identified the p58 subunit of DNA Pol alpha-primase as a new binding partner of XRCC1, a key protein of the single strand break repair (SSBR) complex. In vitro experiments reveal that the binding of poly(ADP-ribose) to p58 inhibits primase activity by competition with its DNA binding property. Overexpression of the XRCC1-BRCT1 domain in HeLa cells induces poly(ADP-ribose) synthesis, PARP-1 and XRCC1-BRCT1 poly(ADP-ribosyl)ation and a strong S phase delay in the presence of DNA damage. Addition of recombinant XRCC1-BRCT1 to Xenopus egg extracts slows down DNA synthesis and inhibits the binding of PCNA, but not MCM2 to alkylated chromatin, thus indicating interference with the assembly of functional replication forks. Altogether these results suggest a critical role for XRCC1 in connecting the SSBR machinery with the replication fork to halt DNA synthesis in response to DNA damage.


Subject(s)
DNA Primase/metabolism , DNA Repair , DNA Replication , DNA-Binding Proteins/metabolism , S Phase/genetics , Animals , Chromatin/metabolism , DNA/biosynthesis , DNA Damage , DNA Polymerase I/metabolism , DNA Primase/chemistry , DNA-Binding Proteins/chemistry , HeLa Cells , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Interaction Domains and Motifs , X-ray Repair Cross Complementing Protein 1 , Xenopus laevis
3.
J Cell Biol ; 167(4): 627-38, 2004 Nov 22.
Article in English | MEDLINE | ID: mdl-15545322

ABSTRACT

The DNA repair proteins poly(ADP-ribose) polymerase-1 (PARP-1), Ku86, and catalytic subunit of DNA-PK (DNA-PKcs) have been involved in telomere metabolism. To genetically dissect the impact of these activities on telomere function, as well as organismal cancer and aging, we have generated mice doubly deficient for both telomerase and any of the mentioned DNA repair proteins, PARP-1, Ku86, or DNA-PKcs. First, we show that abrogation of PARP-1 in the absence of telomerase does not affect the rate of telomere shortening, telomere capping, or organismal viability compared with single telomerase-deficient controls. Thus, PARP-1 does not have a major role in telomere metabolism, not even in the context of telomerase deficiency. In contrast, mice doubly deficient for telomerase and either Ku86 or DNA-PKcs manifest accelerated loss of organismal viability compared with single telomerase-deficient mice. Interestingly, this loss of organismal viability correlates with proliferative defects and age-related pathologies, but not with increased incidence of cancer. These results support the notion that absence of telomerase and short telomeres in combination with DNA repair deficiencies accelerate the aging process without impacting on tumorigenesis.


Subject(s)
Aging, Premature/genetics , Antigens, Nuclear/genetics , Cell Transformation, Neoplastic/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Poly(ADP-ribose) Polymerases/genetics , Protein Serine-Threonine Kinases/genetics , Telomerase/physiology , Aging, Premature/metabolism , Aging, Premature/pathology , Animals , Cell Division/genetics , Cell Transformation, Neoplastic/metabolism , Chromosomal Instability/genetics , DNA-Activated Protein Kinase , DNA-Binding Proteins/deficiency , Female , Ku Autoantigen , Longevity/genetics , Male , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/deficiency , Protein Serine-Threonine Kinases/deficiency , Telomerase/deficiency , Telomerase/genetics , Telomere/genetics
4.
Nucleic Acids Res ; 34(1): 32-41, 2006.
Article in English | MEDLINE | ID: mdl-16397295

ABSTRACT

The two BRCT domains (BRCT1 and BRCT2) of XRCC1 mediate a network of protein-protein interactions with several key factors of the DNA single-strand breaks (SSBs) and base damage repair pathways. BRCT1 is required for the immediate poly(ADP-ribose)-dependent recruitment of XRCC1 to DNA breaks and is essential for survival after DNA damage. To better understand the biological role of XRCC1 in the processing of DNA ends, a search for the BRCT1 domain-associated proteins was performed by mass spectrometry of GST-BRCT1 pulled-down proteins from HeLa cell extracts. Here, we report that the double-strand break (DSB) repair heterotrimeric complex DNA-PK interacts with the BRCT1 domain of XRCC1 and phosphorylates this domain at serine 371 after ionizing irradiation. This caused XRCC1 dimer dissociation. The XRCC1 R399Q variant allele did not affect this phosphorylation. We also show that XRCC1 strongly stimulates the phosphorylation of p53-Ser15 by DNA-PK. The pseudo phosphorylated S371D mutant was a much weaker stimulator of DNA-PK activity whereas the non-phosphorylable mutant S371L endowed with a DNA-PK stimulating capacity failed to fully rescue the DSB repair defect of XRCC1-deficient EM9 rodent cells. The functional association between XRCC1 and DNA-PK in response to IR provides the first evidence for their involvement in a common DSB repair pathway.


Subject(s)
DNA Damage , DNA Repair , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Animals , Binding Sites , Cricetinae , DNA-Binding Proteins/chemistry , Dimerization , HeLa Cells , Humans , Mass Spectrometry , Phosphorylation , Protein Structure, Tertiary , Radiation, Ionizing , Serine/metabolism , X-ray Repair Cross Complementing Protein 1
5.
BMC Mol Biol ; 8: 29, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17459151

ABSTRACT

ATM and PARP-1 are two of the most important players in the cell's response to DNA damage. PARP-1 and ATM recognize and bound to both single and double strand DNA breaks in response to different triggers. Here we report that ATM and PARP-1 form a molecular complex in vivo in undamaged cells and this association increases after gamma-irradiation. ATM is also modified by PARP-1 during DNA damage. We have also evaluated the impact of PARP-1 absence or inhibition on ATM-kinase activity and have found that while PARP-1 deficient cells display a defective ATM-kinase activity and reduced gamma-H2AX foci formation in response to gamma-irradiation, PARP inhibition on itself is able to activate ATM-kinase. PARP inhibition induced gamma H2AX foci accumulation, in an ATM-dependent manner. Inhibition of PARP also induces DNA double strand breaks which were dependent on the presence of ATM. As consequence ATM deficient cells display an increased sensitivity to PARP inhibition. In summary our results show that while PARP-1 is needed in the response of ATM to gamma irradiation, the inhibition of PARP induces DNA double strand breaks (which are resolved in and ATM-dependent pathway) and activates ATM kinase.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adenosine Diphosphate/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line , DNA-Binding Proteins/genetics , Humans , Mice , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics , Protein Binding , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics
6.
Mol Cell Biol ; 24(4): 1595-607, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14749375

ABSTRACT

The DNA damage-dependent poly(ADP-ribose) polymerase-2 (PARP-2) is, together with PARP-1, an active player of the base excision repair process, thus defining its key role in genome surveillance and protection. Telomeres are specialized DNA-protein structures that protect chromosome ends from being recognized and processed as DNA strand breaks. In mammals, telomere protection depends on the T(2)AG(3) repeat binding protein TRF2, which has been shown to remodel telomeres into large duplex loops (t-loops). In this work we show that PARP-2 physically binds to TRF2 with high affinity. The association of both proteins requires the N-terminal domain of PARP-2 and the myb domain of TRF2. Both partners colocalize at promyelocytic leukemia bodies in immortalized telomerase-negative cells. In addition, our data show that PARP activity regulates the DNA binding activity of TRF2 via both a covalent heteromodification of the dimerization domain of TRF2 and a noncovalent binding of poly(ADP-ribose) to the myb domain of TRF2. PARP-2(-/-) primary cells show normal telomere length as well as normal telomerase activity compared to wild-type cells but display a spontaneously increased frequency of chromosome and chromatid breaks and of ends lacking detectable T(2)AG(3) repeats. Altogether, these results suggest a functional role of PARP-2 activity in the maintenance of telomere integrity.


Subject(s)
Poly(ADP-ribose) Polymerases/metabolism , Telomeric Repeat Binding Protein 2/antagonists & inhibitors , Telomeric Repeat Binding Protein 2/metabolism , Animals , Binding Sites , Cell Line , Chromatids/metabolism , Chromosomes, Mammalian/metabolism , DNA/antagonists & inhibitors , DNA/metabolism , DNA Damage , Fibroblasts , Gene Deletion , Humans , Mice , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics , Protein Binding , Protein Structure, Tertiary , Protein Transport , Telomerase/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/chemistry
7.
Methods Enzymol ; 409: 493-510, 2006.
Article in English | MEDLINE | ID: mdl-16793420

ABSTRACT

Changes in chromatin structure emanating from DNA breaks are among the most initiating events in the damage response of the cell. In higher eukaryotes, poly(ADP-ribose) polymerase-1 (PARP-1) translates the occurrence of DNA breaks detected by its zinc-finger domain into a signal, poly ADP-ribose, synthesized and amplified by its DNA-damage dependent catalytic domain. This epigenetic mark on chromatin, induced by DNA discontinuities, is now considered as a part of a survival program aimed at protecting primarily chromatin integrity and stability. In this chapter we describe some of our methods for determining in vivo and in vitro PARP-1 activation in response to DNA strand breaks. Poly(ADP-ribosyl)ation is a posttranslational modification of nuclear proteins induced by DNA strand-breaks that contributes to the survival of injured proliferating cells (D'Amours et al., 1999). Poly(ADP-ribose) polymerases (PARPs) now constitute a large family of 18 proteins, encoded by different genes and displaying a conserved catalytic domain in which PARP-1 (113 kDa), the founding member, and PARP-2 (62 kDa) are so far the sole enzymes whose catalytic activity is immediately stimulated by DNA strand-breaks (Ame et al., 2004). PARP-1 fulfils several key functions in repairing an interruption of the sugar phosphate backbone. It efficiently detects the presence of a break by its N-terminal zinc-finger domain; the occurrence of a break is immediately translated into a posttranslational modification of histones H1 and H2B leading to chromatin structure relaxation and therefore to increased DNA accessibility. As an amplified DNA damage signal, auto-poly(ADP-ribosyl)ation of PARP-1 triggers the recruitment of XRCC1, which coordinates and stimulates the repair process, to the DNA damage sites in less than 15 s in living cells (Okano et al., 2003). Although dispensable in a test tube DNA repair experiment, in vivo these three properties positively influence the overall kinetics of a DNA damage-detection/signaling pathway leading rapidly to the resolution of DNA breaks. Accordingly, poly ADP-ribose (PAR) synthesis and the accompanying NAD consumption are now considered as bona fide marks of DNA interruptions in the genome. In this chapter we describe several methods for determining PARP activation in response to the occurrence of DNA breaks in vitro and in vivo.


Subject(s)
DNA Damage , DNA Repair , Poly(ADP-ribose) Polymerases/metabolism , Animals , Base Sequence , Cell Line , Chromatography, Affinity , DNA Primers , Enzyme Activation , Humans , Mice , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/isolation & purification , Spodoptera
8.
Biochem J ; 386(Pt 1): 119-25, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15456408

ABSTRACT

p53 deficiency confers resistance to doxo (doxorubicin), a clinically active and widely used antitumour anthracycline antibiotic. The purpose of the present study was to investigate the reversal mechanism of doxo resistance by the potent PARP [poly(ADP-ribose) polymerase] inhibitor ANI (4-amino-1,8-naphthalimide) in the p53-deficient breast cancer cell lines EVSA-T and MDA-MB-231. The effects of ANI, in comparison with doxo alone, on doxo-induced apoptosis, were investigated in matched pairs of EVSA-T or MDA-MB-231 with or without ANI co-treatment. Doxo elicited PARP activation as determined by Western blotting and immunofluorescence of poly(ADP-ribose), and ANI enhanced the cytotoxic activity of doxo 2.3 times and in a caspase-dependent manner. The long-term cytotoxic effect was studied by a colony-forming assay. Using this assay, ANI also significantly potentiates the long-term cytotoxic effect with respect to treatment with doxo alone. Decrease in mitochondrial potential together with an increase in cytochrome c release, association of Bax with the mitochondria and caspase 3 activation were also observed in the presence of ANI. Therefore PARP inhibition may represent a novel way of selectively targeting p53-deficient breast cancer cells. The underlying mechanism is probably a potentiation of unrepaired DNA damage, shifting from DNA repair to apoptosis due to the effective inhibition of PARP activity.


Subject(s)
1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplasm Proteins/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors , Quinolones/pharmacology , Tumor Suppressor Protein p53/deficiency , Breast Neoplasms/genetics , Caspase 3 , Caspases/metabolism , Drug Synergism , Female , Genes, p53 , Humans , Intracellular Membranes/drug effects , Membrane Potentials/drug effects , Mitochondria/drug effects , Naphthalimides , Neoplasm Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Stem Cell Assay , bcl-2-Associated X Protein
9.
Nucleic Acids Res ; 31(17): e104, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12930978

ABSTRACT

DNA single strand breaks (SSBs) are one of the most frequent DNA lesions in genomic DNA generated either by oxidative stress or during the base excision repair pathways. Here we established a new real-time assay to assess an imbalance of DNA SSB repair by indirectly measuring PARP-1 activation through the depletion of intracellular NAD(P)H. A water-soluble tetrazolium salt is used to monitor the amount of NAD(P)H in living cells through its reduction to a yellow colored water-soluble formazan dye. While this assay is not a direct method, it does not require DNA extraction or alkaline treatment, both of which could potentially cause an artifactual induction of SSBs. In addition, it takes only 4 h and requires less than a half million cells to perform this measurement. Using this assay, we demonstrated that the dose- and time-dependent depletion of NAD(P)H in XRCC1-deficient CHO cells exposed to methyl methanesulfonate. This decrease was almost completely blocked by a PARP inhibitor. Furthermore, methyl methanesulfonate reduced NAD(P)H in PARP-1+/+ cells, whereas PARP-1-/- cells were more resistant to the decrease in NAD(P)H. These results indicate that the analysis of intracellular NAD(P)H level using water-soluble tetrazolium salt can assess an imbalance of SSB repair in living cells in real time.


Subject(s)
DNA Damage , DNA Repair , DNA, Single-Stranded/genetics , DNA-Binding Proteins/metabolism , NADP/metabolism , Animals , Benzamides/pharmacology , CHO Cells , Comet Assay , Cricetinae , DNA, Single-Stranded/drug effects , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Genotype , Isoquinolines/pharmacology , Methyl Methanesulfonate/pharmacology , Mutation , NAD/metabolism , NADP/drug effects , Piperidines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Time Factors , X-ray Repair Cross Complementing Protein 1
10.
Nucleic Acids Res ; 32(2): 456-64, 2004.
Article in English | MEDLINE | ID: mdl-14739238

ABSTRACT

Poly(ADP-ribose) polymerase-1 (PARP-1) has become an important pharmacological target in the treatment of cancer due to its cellular role as a 'DNA-strand break sensor', which leads in part to resistance to some existing chemo- and radiological treatments. Inhibitors have now been developed which prevent PARP-1 from synthesizing poly(ADP-ribose) in response to DNA-breaks and potentiate the cytotoxicity of DNA damaging agents. However, with the recent discoveries of PARP-2, which has a similar DNA-damage dependent catalytic activity, and additional members containing the 'PARP catalytic' signature, the isoform selectivity and resultant pharmacological effects of existing inhibitors are brought into question. We present here the crystal structure of the catalytic fragment of murine PARP-2, at 2.8 A resolution, and compare this to the catalytic fragment of PARP-1, with an emphasis on providing a possible framework for rational drug design in order to develop future isoform-specific inhibitors.


Subject(s)
Catalytic Domain , Poly(ADP-ribose) Polymerases/chemistry , Amino Acid Sequence , Animals , Binding Sites , Catalysis , Cell Line , Chickens , Crystallization , Crystallography, X-Ray , DNA Damage , Drug Design , Enzyme Inhibitors/chemistry , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Conformation , Substrate Specificity
11.
Nucleic Acids Res ; 31(21): 6198-205, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14576306

ABSTRACT

Poly(ADP-ribose) polymerase-1 (PARP-1) is a key enzyme mediating the cellular response to DNA strand breaks. It plays a critical role in genomic stability and survival of proliferating cells in culture undergoing DNA damage. Intestinal epithelium is the most proliferative tissue in the mammalian body and its stem cells show extreme sensitivity to low-level genotoxic stress. We investigated the role of PARP-1 in the in vivo damage response of intestinal stem cells in crypts of PARP-1-/- and control mice following whole-body gamma-irradiation (1 Gy). In the PARP-1-/- mice there was a significant delay during the first 6 h in the transient p53 accumulation in stem cells whereas an increased number of cells were positive for p21(CIP1/WAF1). Either no or only marginal differences were noted in MDM2 expression, apoptosis, induction of or recovery from mitotic blockage, or inhibition of DNA synthesis. We further observed a dose-dependent reduction in crypt survival measured at 4 days post-irradiation in control mice, and this crypt-killing effect was significantly potentiated in PARP-1-/- mice. Our results thus establish that PARP-1 acts as a survival factor for intestinal stem cells in vivo and suggest a functional link with early p53 and p21(CIP1/WAF1) responses.


Subject(s)
Epithelial Cells/radiation effects , Intestines/radiation effects , Nuclear Proteins , Poly(ADP-ribose) Polymerases/metabolism , Stem Cells/radiation effects , Animals , Apoptosis/radiation effects , Cell Survival/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA Replication/radiation effects , Epithelial Cells/cytology , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Gamma Rays , Gene Deletion , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/enzymology , Mice , Mice, Knockout , Mitosis/radiation effects , Poly(ADP-ribose) Polymerases/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Stem Cells/cytology , Stem Cells/enzymology , Stem Cells/metabolism , Time Factors , Tumor Suppressor Protein p53/metabolism
12.
Oncogene ; 21(7): 1108-16, 2002 Feb 07.
Article in English | MEDLINE | ID: mdl-11850828

ABSTRACT

The tumour suppressor protein p53 plays a key role in the cell's decision to arrest the cell cycle or undergo apoptosis following a genotoxic insult. p53 is stabilized and activated after DNA damage, however the cascade of events signalling from DNA lesions to p53 stabilization and activation is still controversial. Poly (ADP-ribosylation) of different nuclear acceptors by PARP-1 is an early event when a single strand DNA lesion is produced. We present here evidences that interplay between PARP-1 and p53 is dependent on the type of damage induced to DNA. Primary mouse embryonic fibroblasts derived from parp-1 -/- mice exhibited decreased p53 accumulation and activation following gamma-irradiation compared to parp-1 proficient cells. On the other hand, treatment with the single alkylating agent 2'-methyl-2'-nitrose-urea (MNU), resulted in the rapid and sustained accumulation and activation of p53 in parp-1-deficient cells, while very little accumulation was observed in parp-1 +/+ cells. After IR, the turnover of the p53 inhibitory protein MDM-2 is perturbed and the level of phosphorylation of p53 at serine-15 is blunted in parp-1 -/- cells. PARP-1 is determinant in the cytotoxic response to alkylating agents but only partially contributes to radiation-induced cell killing, as determined by colony forming assay. Altogether, these results suggest that PARP-1 participates in the p53 response following irradiation, resides upstream of p53 and indirectly modulates the level of phosphorylation of key substrates in this pathway while treatment with MNU results in an enhanced p53-mediated response in parp-1-null cells.


Subject(s)
DNA Damage , Nuclear Proteins , Poly(ADP-ribose) Polymerases/physiology , Tumor Suppressor Protein p53/physiology , Alkylating Agents/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Gamma Rays , Kinetics , Methylnitrosourea/pharmacology , Mice , Mice, Knockout , Microscopy, Fluorescence , Models, Biological , Poly(ADP-ribose) Polymerases/genetics , Protein Processing, Post-Translational , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , RNA, Messenger/biosynthesis
13.
DNA Repair (Amst) ; 3(8-9): 1103-8, 2004.
Article in English | MEDLINE | ID: mdl-15279798

ABSTRACT

Poly(ADP-ribosyl)ation is an immediate DNA damage-dependent posttranslational modification of histones and nuclear proteins that contributes to the survival of injured proliferating cells. Poly(ADP-ribose) polymerases (PARPs) now constitute a superfamily of 18 proteins, encoded by different genes and displaying a common conserved catalytic domain. PARP-1 (113kDa), the founding member, and PARP-2 (62kDa) are both involved in DNA-break sensing and signaling when single strand break repair (SSBR) or base excision repair (BER) pathways are engaged. The generation by homologous recombination of deficient mouse models have confirmed the caretaker function of PARP-1 and PARP-2 in mammalian cells under genotoxic stress. This review summarizes our present knowledge on their physiological role in the cellular response to DNA damage and on the genetic interactions between PARP-1, PARP-2, Atm that play an essential role during early embryogenesis.


Subject(s)
DNA Damage , Gene Expression Regulation, Developmental , Poly(ADP-ribose) Polymerases/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Catalytic Domain , Cell Cycle Proteins , Cell Proliferation , DNA Repair , DNA-Binding Proteins , Heterozygote , Histones/metabolism , Humans , Mice , Models, Biological , Oxidative Stress , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins
14.
DNA Repair (Amst) ; 2(2): 151-8, 2003 Feb 03.
Article in English | MEDLINE | ID: mdl-12531386

ABSTRACT

Ku is an abundant heterodimeric nuclear protein, consisting of 70- and 86-kDa tightly associated subunits that comprise the DNA binding component of DNA-dependent protein kinase. Poly(ADP-ribose) polymerase-1 (PARP-1) is a 113-kDa protein that catalyzes the synthesis of poly(ADP-ribose) on target proteins. Both Ku and PARP-1 recognize and bind to DNA ends. Ku functions in the non-homologous end joining (NHEJ) repair pathway whereas PARP-1 functions in the single strand break repair and base excision repair (BER) pathways. Recent studies have revealed that PARP-1 and Ku80 interact in vitro. To determine whether the association of PARP-1 and Ku80 has any physiological significance or synergistic function in vivo, mice lacking both PARP-1 and Ku80 were generated. The resulting offspring died during embryonic development displaying abnormalities around the gastrulation stage. In addition, PARP-1-/-/Ku80-/- cultured blastocysts had an increased level of apoptosis. These data suggest that the functions of both Ku80 and PARP-1 are essential for normal embryogenesis and that a loss of genomic integrity leading to cell death through apoptosis is likely the cause of the embryonic lethality observed in these mice.


Subject(s)
Antigens, Nuclear/genetics , DNA Helicases , DNA-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Genes, Lethal , Poly(ADP-ribose) Polymerases/genetics , Animals , Antigens, Nuclear/metabolism , Apoptosis/genetics , Apoptosis/physiology , DNA Repair/genetics , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Ku Autoantigen , Mice , Poly(ADP-ribose) Polymerases/metabolism
15.
J Cell Sci ; 122(Pt 12): 1990-2002, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19454480

ABSTRACT

Poly(ADP-ribosyl)ation is a post-translational modification of proteins involved in the regulation of chromatin structure, DNA metabolism, cell division and cell death. Through the hydrolysis of poly(ADP-ribose) (PAR), Poly(ADP-ribose) glycohydrolase (PARG) has a crucial role in the control of life-and-death balance following DNA insult. Comprehension of PARG function has been hindered by the existence of many PARG isoforms encoded by a single gene and displaying various subcellular localizations. To gain insight into the function of PARG in response to irradiation, we constitutively and stably knocked down expression of PARG isoforms in HeLa cells. PARG depletion leading to PAR accumulation was not deleterious to undamaged cells and was in fact rather beneficial, because it protected cells from spontaneous single-strand breaks and telomeric abnormalities. By contrast, PARG-deficient cells showed increased radiosensitivity, caused by defects in the repair of single- and double-strand breaks and in mitotic spindle checkpoint, leading to alteration of progression of mitosis. Irradiated PARG-deficient cells displayed centrosome amplification leading to mitotic supernumerary spindle poles, and accumulated aberrant mitotic figures, which induced either polyploidy or cell death by mitotic catastrophe. Our results suggest that PARG could be a novel potential therapeutic target for radiotherapy.


Subject(s)
Glycoside Hydrolases/genetics , Mitosis/radiation effects , Radiation Tolerance/genetics , Centrosome/physiology , Centrosome/radiation effects , Chromosome Aberrations/radiation effects , DNA Breaks/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Gene Knockdown Techniques , Glycoside Hydrolases/antagonists & inhibitors , HeLa Cells , Humans , Kinetochores/physiology , Kinetochores/radiation effects , Mitosis/genetics , Poly Adenosine Diphosphate Ribose/metabolism , RNA, Small Interfering/pharmacology , Telomere/radiation effects
16.
Autophagy ; 5(1): 61-74, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19001878

ABSTRACT

Autophagy is a lysosome-dependent degradative pathway frequently activated in tumor cells treated with chemotherapy or radiation. PARP-1 has been implicated in different pathways leading to cell death and its inhibition potentiates chemotherapy-induced cell death. Whether PARP-1 participates in the cell's decision to commit to autophagy following DNA damage is still not known. To address this issue PARP-1 wild-type and deficient cells have been treated with a dose of doxorubicin that induces autophagy. Electron microscopy examination and GFP-LC3 transfection revealed autophagic vesicles and increased expression of genes involved in autophagy (bnip-3, cathepsin b and l and beclin-1) in wild-type cells treated with doxo but not in parp-1(-/-) cells or cells treated with a PARP inhibitor. Mechanistically the lack of autophagic features in PARP-1 deficient/PARP inhibited cells is attributed to prevention of ATP and NAD(+) depletion and to the activation of the key autophagy regulator mTOR. Pharmacological or genetical inhibition of autophagy results in increased cell death, suggesting a protective role of autophagy induced by doxorubicin. These results suggest that autophagy might be cytoprotective during the response to DNA damage and suggest that PARP-1 activation is involved in the cell's decision to undergo autophagy.


Subject(s)
Autophagy , DNA Damage , Poly(ADP-ribose) Polymerases/metabolism , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/pharmacology , 3T3 Cells , Adenosine Triphosphate/deficiency , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Protein 5 , Beclin-1 , Cell Survival/drug effects , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Gene Deletion , Mice , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/ultrastructure , Models, Biological , NAD/deficiency , Naphthalimides/pharmacology , Necrosis/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , Protein Kinases/metabolism , Proteins/metabolism , Quinolones/pharmacology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , TOR Serine-Threonine Kinases , Up-Regulation/drug effects
17.
J Biol Chem ; 282(52): 37738-46, 2007 Dec 28.
Article in English | MEDLINE | ID: mdl-17951580

ABSTRACT

The peroxisome proliferator-activated receptor-gamma (PPARgamma, NR1C3) in complex with the retinoid X receptor (RXR) plays a central role in white adipose tissue (WAT) differentiation and function, regulating the expression of key WAT proteins. In this report we show that poly(ADP-ribose) polymerase-2 (PARP-2), also known as an enzyme participating in the surveillance of the genome integrity, is a member of the PPARgamma/RXR transcription machinery. PARP-2(-/-) mice accumulate less WAT, characterized by smaller adipocytes. In the WAT of PARP-2(-/-) mice the expression of a number of PPARgamma target genes is reduced despite the fact that PPARgamma1 and -gamma2 are expressed at normal levels. Consistent with this, PARP-2(-/-) mouse embryonic fibroblasts fail to differentiate to adipocytes. In transient transfection assays, PARP-2 small interference RNA decreases basal activity and ligand-dependent activation of PPARgamma, whereas PARP-2 overexpression enhances the basal activity of PPARgamma, although it does not change the maximal ligand-dependent activation. In addition, we show a DNA-dependent interaction of PARP-2 and PPARgamma/RXR heterodimer by chromatin immunoprecipitation. In combination, our results suggest that PARP-2 is a novel cofactor of PPARgamma activity.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/physiology , Gene Expression Regulation , PPAR gamma/metabolism , Poly(ADP-ribose) Polymerases/physiology , Retinoid X Receptors/metabolism , 3T3-L1 Cells , Adipose Tissue/metabolism , Animals , Cell Differentiation , Dimerization , Fibroblasts/metabolism , Heterozygote , Mice , Mice, Transgenic , Models, Biological
18.
Nat Rev Mol Cell Biol ; 7(7): 517-28, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16829982

ABSTRACT

The addition to proteins of the negatively charged polymer of ADP-ribose (PAR), which is synthesized by PAR polymerases (PARPs) from NAD(+), is a unique post-translational modification. It regulates not only cell survival and cell-death programmes, but also an increasing number of other biological functions with which novel members of the PARP family have been associated. These functions include transcriptional regulation, telomere cohesion and mitotic spindle formation during cell division, intracellular trafficking and energy metabolism.


Subject(s)
Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Isoforms/metabolism , Animals , Cell Death/physiology , Cell Division/physiology , DNA Damage , DNA Repair , Diphtheria Toxin/chemistry , Humans , Inflammation/metabolism , Models, Molecular , Multigene Family , NAD/biosynthesis , Poly Adenosine Diphosphate Ribose/chemistry , Poly Adenosine Diphosphate Ribose/genetics , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/genetics
19.
Cell Cycle ; 5(8): 873-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16628003

ABSTRACT

Cell survival after genotoxic stress is determined by a counterbalance of pro- and anti-death factors. Sirtuins (SIRTs) are deacetylases that promote cell survival whereas poly(ADP-ribose) polymerases (PARPs) can act both as survival and death inducing factor and the two protein families are strictly dependent on NAD(+) for their activities. Here we report that SIRT1 modulates PARP-1 activity upon DNA damage. Activation of SIRT1 by resveratrol leads to reduced PARP-1 activity and there is a drastic increase in PAR synthesis in sirt1-null cells. The unbalanced regulation of PARP-1 in the absence of SIRT1 results in AIF (apoptosis inducing factor)-mediated cell death. Our findings establish a functional link between the two NAD+-dependent enzyme systems and provide a physiological interpretation for the mechanism of death in cells lacking SIRT1.


Subject(s)
Apoptosis Inducing Factor/physiology , DNA Damage , Gene Expression Regulation , Poly(ADP-ribose) Polymerases/physiology , Sirtuins/physiology , Animals , Cell Death , Cell Line , Enzyme Inhibitors/pharmacology , Humans , Mice , Models, Biological , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Resveratrol , Sirtuin 1 , Stilbenes/pharmacology
20.
Proc Natl Acad Sci U S A ; 103(40): 14854-9, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-17001008

ABSTRACT

Besides the established central role of poly(ADP-ribose) polymerase-1 (Parp-1) and Parp-2 in the maintenance of genomic integrity, accumulating evidence indicates that poly(ADP-ribosyl)ation may modulate epigenetic modifications under physiological conditions. Here, we provide in vivo evidence for the pleiotropic involvement of Parp-2 in both meiotic and postmeiotic processes. We show that Parp-2-deficient mice exhibit severely impaired spermatogenesis, with a defect in prophase of meiosis I characterized by massive apoptosis at pachytene and metaphase I stages. Although Parp-2(-/-) spermatocytes exhibit normal telomere dynamics and normal chromosome synapsis, they display defective meiotic sex chromosome inactivation associated with derailed regulation of histone acetylation and methylation and up-regulated X- and Y-linked gene expression. Furthermore, a drastically reduced number of crossover-associated Mlh1 foci are associated with chromosome missegregation at metaphase I. Moreover, Parp-2(-/-) spermatids are severely compromised in differentiation and exhibit a marked delay in nuclear elongation. Altogether, our findings indicate that, in addition to its well known role in DNA repair, Parp-2 exerts essential functions during meiosis I and haploid gamete differentiation.


Subject(s)
Meiosis/physiology , Poly(ADP-ribose) Polymerases/metabolism , Spermatogenesis/physiology , Animals , Apoptosis , Chromosome Segregation/genetics , Chromosomes, Mammalian/genetics , Infertility, Male , Male , Metaphase/physiology , Mice , Poly(ADP-ribose) Polymerases/deficiency , Sex Chromosomes/genetics , Spermatocytes/cytology , Telomere/metabolism , Testis/cytology
SELECTION OF CITATIONS
SEARCH DETAIL