Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Int J Mol Sci ; 21(14)2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32679873

ABSTRACT

Poly-(ADP-ribosyl)-ation (PARylation) is a reversible post-translational modification of proteins and DNA that plays an important role in various cellular processes such as DNA damage response, replication, transcription, and cell death. Here we designed a fully genetically encoded fluorescent sensor for poly-(ADP-ribose) (PAR) based on Förster resonance energy transfer (FRET). The WWE domain, which recognizes iso-ADP-ribose internal PAR-specific structural unit, was used as a PAR-targeting module. The sensor consisted of cyan Turquoise2 and yellow Venus fluorescent proteins, each in fusion with the WWE domain of RNF146 E3 ubiquitin ligase protein. This bipartite sensor named sPARroW (sensor for PAR relying on WWE) enabled monitoring of PAR accumulation and depletion in live mammalian cells in response to different stimuli, namely hydrogen peroxide treatment, UV irradiation and hyperthermia.


Subject(s)
Bacterial Proteins/analysis , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/analysis , Luminescent Proteins/analysis , Poly Adenosine Diphosphate Ribose/analysis , Bacterial Proteins/genetics , Biosensing Techniques/methods , Cell Line , Fluorescent Dyes/metabolism , Humans , Luminescent Proteins/genetics , Open Reading Frames , Protein Domains , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Ubiquitin-Protein Ligases/analysis , Ubiquitin-Protein Ligases/genetics
2.
Mol Cell Probes ; 39: 57-60, 2018 06.
Article in English | MEDLINE | ID: mdl-29627626

ABSTRACT

Poly (ADP-ribose) polymerase 1 (PARP1) is a DNA damage sensor that catalyzes the poly (ADP-ribose) (PAR) onto a variety of target proteins, such as histones, DSB repair factors and PARP1 itself under consumption of NAD+. Besides, PARP1 can affect a variety of proteins in noncovalent modification manner to carry out specific cellular functions. Here, we established a method to generate non-radiolabeled free PAR by PARG moderately cleaving PAR from autoPARylated PARP1, and utilized dot-blot assay to determine the interaction between free PAR and interested proteins. The methods to generate free PAR and detect the noncovalent interactions between proteins and free PAR are nonradioactive and convenient, which will facilitate the studies to explore the significance of PAR reading in various biological processes.


Subject(s)
Molecular Probe Techniques , Poly Adenosine Diphosphate Ribose/analysis , ELAV-Like Protein 1/metabolism , Humans , Recombinant Fusion Proteins/isolation & purification
3.
Methods Mol Biol ; 1608: 3-18, 2017.
Article in English | MEDLINE | ID: mdl-28695499

ABSTRACT

Poly(ADP-ribosyl)ation (PARylation), i.e., the formation of the nucleic acid-like biopolymer poly(ADP-ribose) (PAR), is an essential posttranslational modification carried out by poly(ADP-ribose) polymerases (PARPs). While PAR levels are low under physiological conditions, they can transiently increase more than 100-fold upon induction of genotoxic stress. The accurate quantitation of cellular PAR with high sensitivity is of critical importance to understand the role of PARylation in cellular physiology and pathophysiology and to determine the pharmacodynamic efficiencies of clinically relevant PARP inhibitors, which represent a novel class of promising chemotherapeutics. Previously, we have developed a bioanalytical platform based on isotope dilution mass spectrometry (LC-MS/MS) to quantify cellular PAR with unequivocal chemical specificity in absolute terms with femtomol sensitivity (Martello et al. ACS Chem Biol 8(7):1567-1575, 2013). This method enables the analysis of steady-state levels, as well as stress-induced levels of PAR in various biological systems including cell lines, mouse tissues, and primary human lymphocytes. It has a wide range of potential applications in basic research, as well as in drug development (Martello et al. ACS Chem Biol 8(7):1567-1575, 2013; Mangerich et al. Toxicol Lett 244:56-71, 2016). Here, we present an improved and adjusted version of the original protocol by Martello/Mangerich et al., which uses UPLC-MS/MS instrumentation.


Subject(s)
Mass Spectrometry/methods , Poly Adenosine Diphosphate Ribose/analysis , Animals , Cell Line , Chromatography, Liquid , DNA Damage/drug effects , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice , Poly Adenosine Diphosphate Ribose/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Tandem Mass Spectrometry
4.
J Surg Res ; 201(2): 313-25, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27020813

ABSTRACT

BACKGROUND: Lung dysfunction constitutes a severe complication after major cardiac surgery with cardiopulmonary bypass (CPB), substantially contributing to postoperative morbidity and mortality. The current possibilities of preventive and therapeutic interventions, however, remain insufficient. We, therefore, investigated the effects of intraoperative application of the antioxidant and anti-inflammatory green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG) on CPB-associated lung injury. MATERIALS AND METHODS: Thirty piglets (8-15 kg) were divided into four groups: sham-operated and saline-treated control group (n = 7); sham-operated and EGCG-treated control group (EGCG-control group; n = 7); CPB group (n = 10); and CPB + EGCG group (n = 6). The CPB groups underwent 120 min of CPB followed by 90 min of recovery time. In the CPB + EGCG group, EGCG (10 mg/kg body weight) was administered intravenously before and after CPB. Hemodynamic monitoring, blood gas analysis, hematoxylin-eosin staining, and immunohistochemistry of lung tissue were performed. RESULTS: Histologic examination revealed thickening of the alveolar wall and enhanced alveolar neutrophil infiltration in the CPB group (P < 0.05) compared with those in the control group, which was prevented by EGCG (P < 0.05). In the CPB group, higher formation of poly(ADP-ribose) and nuclear translocation of apoptosis-inducing factor was detected in comparison with those in the control group (P < 0.001), which were both reduced in the CPB + EGCG group (P < 0.001). Compared with the control group, the EGCG-control group showed thickening of the alveolar wall and increased neutrophil infiltration (P < 0.05). CONCLUSIONS: CPB leads to lung edema, pulmonary neutrophil infiltration, and presumably initiation of poly(ADP-ribose) polymerase-dependent cell death signaling in the lung. EGCG appears to attenuate CPB-associated lung injury, suggesting that this may provide a novel pharmacologic approach.


Subject(s)
Antioxidants/therapeutic use , Cardiopulmonary Bypass/adverse effects , Catechin/analogs & derivatives , Lung Injury/prevention & control , Animals , Apoptosis Inducing Factor/analysis , Camellia sinensis , Catechin/therapeutic use , Drug Evaluation, Preclinical , Female , Immunohistochemistry , Lung/chemistry , Lung/pathology , Lung Injury/etiology , Lung Injury/pathology , Male , Phytotherapy , Plant Extracts/therapeutic use , Poly Adenosine Diphosphate Ribose/analysis , Swine , Tumor Necrosis Factor-alpha/analysis , Tyrosine/analogs & derivatives , Tyrosine/analysis
5.
PLoS One ; 11(2): e0148727, 2016.
Article in English | MEDLINE | ID: mdl-26866363

ABSTRACT

Many studies have demonstrated changes in the levels of several ions during apoptosis, but a few recent studies have reported conflicting results concerning the changes in water content in apoptotic cells. We used a correlative light and cryo-scanning transmission electron microscopy method to quantify water and ion/element contents simultaneously at a nanoscale resolution in the various compartments of cells, from the onset to the end of apoptosis. We used stably transfected HeLa cells producing H2B-GFP to identify the stages of apoptosis in cells and for a targeted elemental analysis within condensed chromatin, nucleoplasm, mitochondria and the cytosol. We found that the compartments of apoptotic cells contained, on average, 10% more water than control cells. During mitochondrial outer membrane permeabilization, we observed a strong increase in the Na+ and Cl- contents of the mitochondria and a strong decrease in mitochondrial K+ content. During the first step in apoptotic volume decrease (AVD), Na+ and Cl- levels decreased in all cell compartments, but remained higher than those in control cells. Conversely, during the second step of AVD, Na+ and Cl- levels increased considerably in the nucleus and mitochondria. During these two steps of AVD, K+ content decreased steadily in all cell compartments. We also determined in vivo ion status during caspase-3 activity and chromatin condensation. Finally, we found that actinomycin D-tolerant cells had water and K+ contents similar to those of cells entering apoptosis but lower Na+ and Cl- contents than both cells entering apoptosis and control cells.


Subject(s)
Apoptosis , Chlorides/analysis , Cryoelectron Microscopy/methods , Nanotechnology/methods , Organelles/chemistry , Potassium/analysis , Sodium/analysis , Spectrometry, X-Ray Emission/methods , Time-Lapse Imaging/methods , Water/analysis , Anions/analysis , Apoptosis/drug effects , Caspase 3/analysis , Cations/analysis , Cell Membrane Permeability , Cell Size , Cytochromes c/analysis , Dactinomycin/pharmacology , HeLa Cells , Humans , Image Processing, Computer-Assisted , Microscopy, Electron, Scanning/methods , Mitochondria/chemistry , Mitochondrial Membranes , Poly Adenosine Diphosphate Ribose/analysis
6.
Clin Cancer Res ; 22(12): 2855-64, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26801247

ABSTRACT

PURPOSE: Cisplatin is synergistic with vinorelbine and the PARP inhibitor veliparib, and has antineoplastic activity in triple-negative breast cancer (TNBC) and BRCA mutation-associated breast cancer. This phase I study assessed veliparib with cisplatin and vinorelbine. EXPERIMENTAL DESIGN: A 3+3 dose-escalation design evaluated veliparib administered twice daily for 14 days with cisplatin (75 mg/m(2) day 1) and vinorelbine (25 mg/m(2) days 1, 8) every 21 days, for 6 to 10 cycles, followed by veliparib monotherapy. Pharmacokinetics, measurement of poly(ADP-ribose) in peripheral blood mononuclear cells, and preliminary efficacy were assessed. IHC and gene-expression profiling were evaluated as potential predictors of response. RESULTS: Forty-five patients enrolled in nine dose cohorts plus five in an expansion cohort at the highest dose level and recommended phase II dose, 300 mg twice daily. The MTD of veliparib was not reached. Neutropenia (36%), anemia (30%), and thrombocytopenia (12%) were the most common grade 3/4 adverse events. Best overall response for 48 patients was radiologic response with 9-week confirmation for 17 (35%; 2 complete, 15 partial), and stable disease for 21 (44%). Germline BRCA mutation presence versus absence was associated with 6-month progression-free survival [PFS; 10 of 14 (71%) vs. 8 of 27 (30%), mid-P = 0.01]. Median PFS for all 50 patients was 5.5 months (95% confidence interval, 4.1-6.7). CONCLUSIONS: Veliparib at 300 mg twice daily combined with cisplatin and vinorelbine is well tolerated with encouraging response rates. A phase II randomized trial is planned to assess veliparib's contribution to cisplatin chemotherapy in metastatic TNBC and BRCA mutation-associated breast cancer. Clin Cancer Res; 22(12); 2855-64. ©2016 AACR.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , BRCA2 Protein/genetics , Benzimidazoles/therapeutic use , Cisplatin/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Vinblastine/analogs & derivatives , Adult , Aged , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Cisplatin/adverse effects , Cisplatin/pharmacokinetics , DNA Repair/genetics , Disease-Free Survival , Female , Humans , Middle Aged , Poly Adenosine Diphosphate Ribose/analysis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Vinblastine/adverse effects , Vinblastine/pharmacokinetics , Vinblastine/therapeutic use , Vinorelbine
7.
Anal Biochem ; 494: 76-81, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26548958

ABSTRACT

PolyADP-ribosylation is mediated by poly(ADP-ribose) (PAR) polymerases (PARPs) and may be involved in various cellular events, including chromosomal stability, DNA repair, transcription, cell death, and differentiation. The physiological level of PAR is difficult to determine in intact cells because of the rapid synthesis of PAR by PARPs and the breakdown of PAR by PAR-degrading enzymes, including poly(ADP-ribose) glycohydrolase (PARG) and ADP-ribosylhydrolase 3. Artifactual synthesis and/or degradation of PAR likely occurs during lysis of cells in culture. We developed a sensitive enzyme-linked immunosorbent assay (ELISA) to measure the physiological levels of PAR in cultured cells. We immediately inactivated enzymes that catalyze the synthesis and degradation of PAR. We validated that trichloroacetic acid is suitable for inactivating PARPs, PARG, and other enzymes involved in metabolizing PAR in cultured cells during cell lysis. The PAR level in cells harvested with the standard radioimmunoprecipitation assay buffer was increased by 450-fold compared with trichloroacetic acid for lysis, presumably because of activation of PARPs by DNA damage that occurred during cell lysis. This ELISA can be used to analyze the biological functions of polyADP-ribosylation under various physiological conditions in cultured cells.


Subject(s)
Chemistry Techniques, Analytical/methods , Enzyme-Linked Immunosorbent Assay , Poly Adenosine Diphosphate Ribose/analysis , Antibodies/immunology , DNA Damage , Deoxyribonuclease I/metabolism , Glycoside Hydrolases/metabolism , HEK293 Cells , HeLa Cells , Humans , Poly Adenosine Diphosphate Ribose/immunology , Poly(ADP-ribose) Polymerases/metabolism , Radioimmunoprecipitation Assay , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Trichloroacetic Acid/chemistry
8.
DNA Repair (Amst) ; 30: 68-79, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25800440

ABSTRACT

An important feature of poly(ADP-ribose) polymerases (PARPs) is their ability to readily undergo automodification upon activation. Although a growing number of substrates were found to be poly(ADP-ribosyl)ated, including histones and several DNA damage response factors, PARPs themselves are still considered as the main acceptors of poly(ADP-ribose). By monitoring spectral counts of specific hydroxamic acid signatures generated after the conversion of the ADP-ribose modification onto peptides by hydroxylamine hydrolysis, we undertook a thorough mass spectrometry mapping of the glutamate and aspartate ADP-ribosylation sites onto automodified PARP-1, PARP-2 and PARP-3. Thousands of hydroxamic acid-conjugated peptides were identified with high confidence and ranked based on their spectral count. This semi-quantitative approach allowed us to locate the preferentially targeted residues in DNA-dependent PARPs. In contrast to what has been reported in the literature, automodification of PARP-1 is not predominantly targeted towards its BRCT domain. Our results show that interdomain linker regions that connect the BRCT to the WGR module and the WGR to the PRD domain undergo prominent ADP-ribosylation during PARP-1 automodification. We also found that PARP-1 efficiently automodifies the D-loop structure within its own catalytic fold. Interestingly, additional major ADP-ribosylation sites were identified in functional domains of PARP-1, including all three zinc fingers. Similar to PARP-1, specific residues located within the catalytic sites of PARP-2 and PARP-3 are major targets of automodification following their DNA-dependent activation. Together our results suggest that poly(ADP-ribosyl)ation hot spots make a dominant contribution to the overall automodification process.


Subject(s)
Cell Cycle Proteins/chemistry , Poly Adenosine Diphosphate Ribose/analysis , Poly(ADP-ribose) Polymerases/chemistry , Animals , Cattle , Cell Cycle Proteins/metabolism , Humans , Mass Spectrometry , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Structure, Tertiary
9.
Proteomics ; 15(2-3): 203-17, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25263235

ABSTRACT

ADP-ribosylation refers to the addition of one or more ADP-ribose units onto protein substrates and this protein modification has been implicated in various cellular processes including DNA damage repair, RNA metabolism, transcription, and cell cycle regulation. This review focuses on a compilation of large-scale proteomics studies that identify ADP-ribosylated proteins and their associated proteins by MS using a variety of enrichment strategies. Some methods, such as the use of a poly(ADP-ribose)-specific antibody and boronate affinity chromatography and NAD(+) analogues, have been employed for decades while others, such as the use of protein microarrays and recombinant proteins that bind ADP-ribose moieties (such as macrodomains), have only recently been developed. The advantages and disadvantages of each method and whether these methods are specific for identifying mono(ADP-ribosyl)ated and poly(ADP-ribosyl)ated proteins will be discussed. Lastly, since poly(ADP-ribose) is heterogeneous in length, it has been difficult to attain a mass signature associated with the modification sites. Several strategies on how to reduce polymer chain length heterogeneity for site identification will be reviewed.


Subject(s)
Adenosine Diphosphate Ribose/analysis , Mass Spectrometry/methods , Poly Adenosine Diphosphate Ribose/analysis , Proteins/chemistry , Proteomics/methods , Adenosine Diphosphate Ribose/metabolism , Animals , Humans , NAD/analogs & derivatives , NAD/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Proteins/metabolism
10.
DNA Repair (Amst) ; 26: 23-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25541391

ABSTRACT

Base excision repair (BER) is a primary mechanism for repair of base lesions in DNA such as those formed by exposure to the DNA methylating agent methyl methanesulfonate (MMS). Both DNA polymerase ß (pol ß)- and XRCC1-deficient mouse fibroblasts are hypersensitive to MMS. This is linked to a repair deficiency as measured by accumulation of strand breaks and poly(ADP-ribose) (PAR). The interaction between pol ß and XRCC1 is important for recruitment of pol ß to sites of DNA damage. Endogenous DNA damage can substitute for MMS-induced damage such that BER deficiency as a result of either pol ß- or XRCC1-deletion is associated with sensitivity to PARP inhibitors. Pol ß shRNA was used to knock down pol ß in Xrcc1(+/+) and Xrcc1(-/-) mouse fibroblasts. We determined whether pol ß-mediated cellular resistance to MMS and PARP inhibitors resulted entirely from coordination with XRCC1 within the same BER sub-pathway. We find evidence for pol ß-dependent cell survival independent of XRCC1 expression for both types of agents. The results suggest a role for pol ß-dependent, XRCC1-independent repair. PAR immunofluorescence data are consistent with the hypothesis of a decrease in repair in both pol ß knock down cell variants.


Subject(s)
DNA Polymerase beta/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Alkylating Agents/pharmacology , Animals , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/genetics , DNA/metabolism , DNA Adducts/metabolism , DNA Polymerase beta/genetics , DNA Repair/drug effects , DNA Repair/genetics , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Methyl Methanesulfonate/pharmacology , Mice , Mice, Knockout , Poly Adenosine Diphosphate Ribose/analysis , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/pharmacology , X-ray Repair Cross Complementing Protein 1
11.
Science ; 344(6185): 742-6, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24833391

ABSTRACT

Nuclear magnetic resonance (NMR) spectroscopy is useful to determine molecular structure in tissues grown in vitro only if their fidelity, relative to native tissue, can be established. Here, we use multidimensional NMR spectra of animal and in vitro model tissues as fingerprints of their respective molecular structures, allowing us to compare the intact tissues at atomic length scales. To obtain spectra from animal tissues, we developed a heavy mouse enriched by about 20% in the NMR-active isotopes carbon-13 and nitrogen-15. The resulting spectra allowed us to refine an in vitro model of developing bone and to probe its detailed structure. The identification of an unexpected molecule, poly(adenosine diphosphate ribose), that may be implicated in calcification of the bone matrix, illustrates the analytical power of this approach.


Subject(s)
Bone Development , Calcification, Physiologic , Nuclear Magnetic Resonance, Biomolecular/methods , Poly Adenosine Diphosphate Ribose/analysis , Animals , Carbon Isotopes , Extracellular Matrix/chemistry , Growth Plate/growth & development , Mice , Models, Biological , Nitrogen Isotopes , Sheep
12.
Metallomics ; 6(3): 662-71, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24549367

ABSTRACT

The toxicologically most relevant mercury (Hg) species for human exposure is methylmercury (MeHg). Thiomersal is a common preservative used in some vaccine formulations. The aim of this study is to get further mechanistic insight into the yet not fully understood neurotoxic modes of action of organic Hg species. Mercury species investigated include MeHgCl and thiomersal. Additionally HgCl2 was studied, since in the brain mercuric Hg can be formed by dealkylation of the organic species. As a cellular system astrocytes were used. In vivo astrocytes provide the environment necessary for neuronal function. In the present study, cytotoxic effects of the respective mercuricals increased with rising alkylation level and correlated with their cellular bioavailability. Further experiments revealed for all species at subcytotoxic concentrations no induction of DNA strand breaks, whereas all species massively increased H2O2-induced DNA strand breaks. This co-genotoxic effect is likely due to a disturbance of the cellular DNA damage response. Thus, at nanomolar, sub-cytotoxic concentrations, all three mercury species strongly disturbed poly(ADP-ribosyl)ation, a signalling reaction induced by DNA strand breaks. Interestingly, the molecular mechanism behind this inhibition seems to be different for the species. Since chronic PARP-1 inhibition is also discussed to sacrifice neurogenesis and learning abilities, further experiments on neurons and in vivo studies could be helpful to clarify whether the inhibition of poly(ADP-ribosyl)ation contributes to organic Hg induced neurotoxicity.


Subject(s)
Astrocytes/drug effects , Methylmercury Compounds/toxicity , Mutagens/toxicity , Thimerosal/toxicity , Astrocytes/metabolism , Cell Line , DNA Breaks/drug effects , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/analysis , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism
13.
Mol Cell Biol ; 33(4): 845-57, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230272

ABSTRACT

The BAL1 macrodomain-containing protein and its partner E3 ligase, BBAP, are overexpressed in chemotherapy-resistant lymphomas. BBAP selectively ubiquitylates histone H4 and indirectly promotes early 53BP1 recruitment to DNA damage sites. However, neither BBAP nor BAL1 has been directly associated with a DNA damage response (DDR), and the function of BAL1 remains undefined. Herein, we describe a direct link between rapid and short-lived poly(ADP-ribose) (PAR) polymerase 1 (PARP1) activation and PARylation at DNA damage sites, PAR-dependent recruitment of the BAL1 macrodomain-containing protein and its partner E3 ligase, local BBAP-mediated ubiquitylation, and subsequent recruitment of the checkpoint mediators 53BP1 and BRCA1. The PARP1-dependent localization of BAL1-BBAP functionally limits both early and delayed DNA damage and enhances cellular viability independent of ATM, MDC1, and RNF8. These data establish that BAL1 and BBAP are bona fide members of a DNA damage response pathway and are directly associated with PARP1 activation, BRCA1 recruitment, and double-strand break repair.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/analysis , Carrier Proteins/metabolism , Cell Cycle Proteins/analysis , Cell Line , DNA Repair , DNA-Binding Proteins/analysis , Histone Chaperones , Humans , Neoplasm Proteins/analysis , Nuclear Proteins/analysis , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/analysis , Poly(ADP-ribose) Polymerases/analysis , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/analysis , Protein Structure, Tertiary , Trans-Activators/analysis , Tumor Suppressor Proteins/analysis , Ubiquitin-Protein Ligases/analysis , Ubiquitination
14.
Methods Mol Biol ; 780: 117-34, 2011.
Article in English | MEDLINE | ID: mdl-21870258

ABSTRACT

Poly(ADP-ribose) polymerases (PARP) participate in diverse biological processes contributing to cellular homeostasis or exacerbating injury. PARP catalyzes the addition of ADP-ribose molecules (pADPr) to the target proteins, a process termed poly-ADP-ribosylation. Overactivation of PARP, as reflected by increased poly-ADP-ribosylation, accumulation of pADPr-modified proteins or free pADPr, contributes to the depletion of NAD(+) and mitochondrial dysfunction, potentially leading to cell death via apoptosis or necrosis. Since PARP over-activation has been identified as a key contributor to the pathobiology of many diseases, monitoring PARP 1 activation by detecting and quantifying pADPr may provide valuable mechanistic insights as well as facilitating therapeutic drug monitoring for PARP inhibitors.Several non-isotopic immunodetection methods for quantifying pADPr are discussed: western blotting of poly-ADP-ribosylated proteins, cellular localization of pADPr by immunohistochemistry, quantification of pADPr by enzyme-linked immunoassay and small-scale two-dimensional gel electrophoresis.


Subject(s)
Poly(ADP-ribose) Polymerases/metabolism , Animals , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice , NAD , Poly Adenosine Diphosphate Ribose/analysis , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/analysis , Rats
15.
Analyst ; 136(10): 2044-6, 2011 May 21.
Article in English | MEDLINE | ID: mdl-21448503

ABSTRACT

A new method to assay protein poly(ADP-ribosyl)ation was proposed based on the interaction between the substrate of poly(ADP-ribosyl)ation nicotinamide adenine dinucleotide and gold nanoparticles, which needed no coupled enzymes or other modified catalytic substrate.


Subject(s)
Colorimetry/methods , Gold/chemistry , Metal Nanoparticles/chemistry , Poly Adenosine Diphosphate Ribose/analysis , Proteins/analysis , DNA/chemistry , NAD/chemistry
16.
Chem Commun (Camb) ; 47(1): 397-9, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-20830433

ABSTRACT

Designed sensors comprising split-firefly luciferase conjugated to tandem poly(ADP-ribose) binding domains allow for the direct solution phase detection of picogram quantities of PAR and for monitoring temporal changes in poly(ADP-ribosyl)ation events in mammalian cells.


Subject(s)
Biomarkers, Tumor/analysis , Biosensing Techniques/methods , Luciferases/metabolism , Poly Adenosine Diphosphate Ribose/analysis , Biomarkers, Tumor/metabolism , Cell Death , Cell Line, Tumor , DNA Repair , HeLa Cells , Humans , Luciferases/chemistry , Molecular Conformation , Poly Adenosine Diphosphate Ribose/metabolism
17.
Clin Cancer Res ; 16(18): 4532-42, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20823146

ABSTRACT

Tumor cells are often deficient in DNA damage response (DDR) pathways, and anticancer therapies are commonly based on genotoxic treatments using radiation and/or drugs that damage DNA directly or interfere with DNA metabolism, leading to the formation of DNA double-strand breaks (DSB), and ultimately to cell death. Because DSBs induce the phosphorylation of histone H2AX (γH2AX) in the chromatin flanking the break site, an antibody directed against γH2AX can be employed to measure DNA damage levels before and after patient treatment. Poly(ADP-ribose) polymerases (PARP1 and PARP2) are also activated by DNA damage, and PARP inhibitors show promising activity in cancers with defective homologous recombination (HR) pathways for DSB repair. Ongoing clinical trials are testing combinations of PARP inhibitors with DNA damaging agents. Poly(ADP-ribosylation), abbreviated as PAR, can be measured in clinical samples and used to determine the efficiency of PARP inhibitors. This review summarizes the roles of γH2AX and PAR in the DDR, and their use as biomarkers to monitor drug response and guide clinical trials, especially phase 0 clinical trials. We also discuss the choices of relevant samples for γH2AX and PAR analyses.


Subject(s)
Biomarkers, Pharmacological , Biomarkers, Tumor/physiology , Histones/physiology , Neoplasms/diagnosis , Poly Adenosine Diphosphate Ribose/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Pharmacological/analysis , Biomarkers, Pharmacological/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , DNA Damage/drug effects , DNA Damage/genetics , Histones/genetics , Histones/metabolism , Humans , Medical Oncology/methods , Models, Biological , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Poly Adenosine Diphosphate Ribose/analysis , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/physiology , Prognosis
18.
Crit Care ; 14(4): R129, 2010.
Article in English | MEDLINE | ID: mdl-20602787

ABSTRACT

INTRODUCTION: Different isoforms of nitric oxide synthases (NOS) and determinants of oxidative/nitrosative stress play important roles in the pathophysiology of pulmonary dysfunction induced by acute lung injury (ALI) and sepsis. However, the time changes of these pathogenic factors are largely undetermined. METHODS: Twenty-four chronically instrumented sheep were subjected to inhalation of 48 breaths of cotton smoke and instillation of live Pseudomonas aeruginosa into both lungs and were euthanized at 4, 8, 12, 18, and 24 hours post-injury. Additional sheep received sham injury and were euthanized after 24 hrs (control). All animals were mechanically ventilated and fluid resuscitated. Lung tissue was obtained at the respective time points for the measurement of neuronal, endothelial, and inducible NOS (nNOS, eNOS, iNOS) mRNA and their protein expression, calcium-dependent and -independent NOS activity, 3-nitrotyrosine (3-NT), and poly(ADP-ribose) (PAR) protein expression. RESULTS: The injury induced severe pulmonary dysfunction as indicated by a progressive decline in oxygenation index and concomitant increase in pulmonary shunt fraction. These changes were associated with an early and transient increase in eNOS and an early and profound increase in iNOS expression, while expression of nNOS remained unchanged. Both 3-NT, a marker of protein nitration, and PAR, an indicator of DNA damage, increased early but only transiently. CONCLUSIONS: Identification of the time course of the described pathogenetic factors provides important additional information on the pulmonary response to ALI and sepsis in the ovine model. This information may be crucial for future studies, especially when considering the timing of novel treatment strategies including selective inhibition of NOS isoforms, modulation of peroxynitrite, and PARP.


Subject(s)
Nitric Oxide Synthase/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Sepsis/metabolism , Tyrosine/analogs & derivatives , Acute Lung Injury/metabolism , Acute Lung Injury/physiopathology , Animals , Disease Models, Animal , Interleukin-8/analysis , Interleukin-8/metabolism , Interleukin-8/physiology , Lung/chemistry , Lung/enzymology , Nitrates/blood , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type II/physiology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type III/physiology , Nitrites/blood , Poly Adenosine Diphosphate Ribose/analysis , Poly Adenosine Diphosphate Ribose/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/physiopathology , Sheep , Time Factors , Tyrosine/analysis , Tyrosine/metabolism , Tyrosine/physiology
19.
J Biol Chem ; 284(30): 20175-83, 2009 Jul 24.
Article in English | MEDLINE | ID: mdl-19447888

ABSTRACT

Micro opioid receptor (MOR) agonists such as morphine are applied widely in clinical practice as pain therapy. The effects of morphine through MOR, such as analgesia and development of tolerance and dependence, are influenced by individual specificity. Recently, we analyzed single nucleotide polymorphisms on the human MOR gene to investigate the factors that contribute to individual specificity. In process of single nucleotide polymorphisms analysis, we found that specific nuclear proteins bound to G(-172) --> T region in exon 1 in MOR gene, and its affinity to DNA was increased by base substitution from G(-172) to T(-172). The isolated protein was identified by mass spectrometry and was confirmed by Western blotting to be poly(ADP-ribose) polymerase-1 (PARP-1). The overexpressed PARP-1 bound to G(-172) --> T and enhanced the transcription of reporter vectors containing G(-172) and T(-172). Furthermore, PARP-1 inhibitor (benzamide) decreased PARP-1 binding to G(-172) --> T without affecting mRNA or protein expression level of PARP-1 and down-regulated the subsequent MOR gene expression in SH-SY5Y cells. Moreover, we found that tumor necrosis factor-alpha enhanced MOR gene expression as well as increased PARP-1 binding to the G(-172) --> T region and G(-172) --> T-dependent transcription in SH-SY5Y cells. These effects were also inhibited by benzamide. In this study, our data suggest that PARP-1 positively regulates MOR gene transcription via G(-172) --> T, which might influence individual specificity in therapeutic opioid effects.


Subject(s)
Poly Adenosine Diphosphate Ribose/analysis , Poly Adenosine Diphosphate Ribose/metabolism , Polymorphism, Single Nucleotide , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Tumor Necrosis Factor-alpha/metabolism , Base Sequence , Benzamides/pharmacology , Binding Sites , Cell Line , Cell Line, Tumor , DNA/genetics , DNA/metabolism , Exons , Gene Expression Regulation , Humans , Kidney/cytology , Neuroblastoma/genetics , Neuroblastoma/metabolism , Poly Adenosine Diphosphate Ribose/antagonists & inhibitors , Poly Adenosine Diphosphate Ribose/genetics , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcriptional Activation
20.
J Virol ; 82(4): 1748-58, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18057239

ABSTRACT

Infection of neonatal rats with Borna disease virus results in a characteristic behavioral syndrome and apoptosis of subsets of neurons in the hippocampus, cerebellum, and cortex (neonatal Borna disease [NBD]). In the NBD rat hippocampus, dentate gyrus granule cells progressively degenerate. Apoptotic loss of granule cells in NBD is associated with accumulation of zinc in degenerating neurons and reduced zinc in granule cell mossy fibers. Excess zinc can trigger poly(ADP-ribose) polymerase 1 (PARP-1) activation, and PARP-1 activation can mediate neuronal death. Here, we evaluate hippocampal PARP-1 mRNA and protein expression levels, activation, and cleavage, as well as apoptosis-inducing factor (AIF) nuclear translocation and executioner caspase 3 activation, in NBD rats. PARP-1 mRNA and protein levels were increased in NBD hippocampi. PARP-1 expression and activity were increased in granule cell neurons and glia with enhanced ribosylation of proteins, including PARP-1 itself. In contrast, levels of poly(ADP-ribose) glycohydrolase mRNA were decreased in NBD hippocampi. PARP-1 cleavage and AIF expression were also increased in astrocytes in NBD hippocampi. Levels of activated caspase 3 protein were increased in NBD hippocampi and localized to nuclei, mossy fibers, and dendrites of granule cell neurons. These results implicate aberrant zinc homeostasis, PARP-1, and caspase 3 activation as contributing factors in hippocampal neurodegeneration in NBD.


Subject(s)
Borna Disease/pathology , Caspase 3/metabolism , Hippocampus/pathology , Hippocampus/virology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/virology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Animals, Newborn , Apoptosis , Apoptosis Inducing Factor/analysis , Apoptosis Inducing Factor/metabolism , Borna Disease/enzymology , Caspase 3/analysis , Cerebral Cortex/enzymology , Dentate Gyrus/enzymology , Dentate Gyrus/pathology , Dentate Gyrus/virology , Female , Hippocampus/enzymology , Neurodegenerative Diseases/enzymology , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/analysis , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/analysis , Poly(ADP-ribose) Polymerases/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...