Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Mol Cancer ; 14: 207, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26654227

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is a clinically heterogeneous lymphoid malignancy and the most common subtype of non-Hodgkin's lymphoma in adults, with one of the highest mortality rates in most developed areas of the world. More than half of DLBLC patients can be cured with standard R-CHOP regimens, however approximately 30 to 40 % of patients will develop relapsed/refractory disease that remains a major cause of morbidity and mortality due to the limited therapeutic options.Recent advances in gene expression profiling have led to the identification of at least three distinct molecular subtypes of DLBCL: a germinal center B cell-like subtype, an activated B cell-like subtype, and a primary mediastinal B-cell lymphoma subtype. Moreover, recent findings have not only increased our understanding of the molecular basis of chemotherapy resistance but have also helped identify molecular subsets of DLBCL and rational targets for drug interventions that may allow for subtype/subset-specific molecularly targeted precision medicine and personalized combinations to both prevent and treat relapsed/refractory DLBCL. Novel agents such as lenalidomide, ibrutinib, bortezomib, CC-122, epratuzumab or pidilizumab used as single-agent or in combination with (rituximab-based) chemotherapy have already demonstrated promising activity in patients with relapsed/refractory DLBCL. Several novel potential drug targets have been recently identified such as the BET bromodomain protein (BRD)-4, phosphoribosyl-pyrophosphate synthetase (PRPS)-2, macrodomain-containing mono-ADP-ribosyltransferase (ARTD)-9 (also known as PARP9), deltex-3-like E3 ubiquitin ligase (DTX3L) (also known as BBAP), NF-kappaB inducing kinase (NIK) and transforming growth factor beta receptor (TGFßR).This review highlights the new insights into the molecular basis of relapsed/refractory DLBCL and summarizes the most promising drug targets and experimental treatments for relapsed/refractory DLBCL, including the use of novel agents such as lenalidomide, ibrutinib, bortezomib, pidilizumab, epratuzumab, brentuximab-vedotin or CAR T cells, dual inhibitors, as well as mechanism-based combinatorial experimental therapies. We also provide a comprehensive and updated list of current drugs, drug targets and preclinical and clinical experimental studies in DLBCL. A special focus is given on STAT1, ARTD9, DTX3L and ARTD8 (also known as PARP14) as novel potential drug targets in distinct molecular subsets of DLBCL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, Large B-Cell, Diffuse/drug therapy , Neoplasm Recurrence, Local/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm , Humans , Immunotherapy , Lymphoma, Large B-Cell, Diffuse/pathology , Molecular Targeted Therapy , Precision Medicine
3.
Mol Cancer ; 13: 125, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24886089

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is one of the leading causes of cancer-related mortality and morbidity in the aging male population and represents the most frequently diagnosed malignancy in men around the world. The Deltex (DTX)-3-like E3 ubiquitin ligase (DTX3L), also known as B-lymphoma and BAL-associated protein (BBAP), was originally identified as a binding partner of the diphtheria-toxin-like macrodomain containing ADP-ribosyltransferase-9 (ARTD9), also known as BAL1 and PARP9. We have previously demonstrated that ARTD9 acts as a novel oncogenic survival factor in high-risk, chemo-resistant, diffuse large B cell lymphoma (DLBCL). The mono-ADP-ribosyltransferase ARTD8, also known as PARP14 functions as a STAT6-specific co-regulator of IL4-mediated proliferation and survival in B cells. METHODS: Co-expression of DTX3L, ARTD8, ARTD9 and STAT1 was analyzed in the metastatic PCa (mPCa) cell lines PC3, DU145, LNCaP and in the normal prostate luminal epithelial cell lines HPE and RWPE1. Effects on cell proliferation, survival and cell migration were determined in PC3, DU145 and/or LNCaP cells depleted of DTX3L, ARTD8, ARTD9, STAT1 and/or IRF1 compared to their proficient control cells, respectively. In further experiments, real-time RT-PCR, Western blot, immunofluorescence and co-immunoprecipitations were conducted to evaluate the physical and functional interactions between DTX3L, ARTD8 and ARTD9. RESULTS: Here we could identify DTX3L, ARTD9 and ARTD8 as novel oncogenic survival factors in mPCa cells. Our studies revealed that DTX3L forms a complex with ARTD8 and mediates together with ARTD8 and ARTD9 proliferation, chemo-resistance and survival of mPCa cells. In addition, DTX3L, ARTD8 and ARTD9 form complexes with each other. Our study provides first evidence that the enzymatic activity of ARTD8 is required for survival of mPCa cells. DTX3L and ARTD9 act together as repressors of the tumor suppressor IRF1 in mPCa cells. Furthermore, the present study shows that DTX3L together with STAT1 and STAT3 is implicated in cell migration of mPCa cells. CONCLUSIONS: Our data strongly indicate that a crosstalk between STAT1, DTX3L and ARTD-like mono-ADP-ribosyltransferases mediates proliferation and survival of mPCa cells. The present study further suggests that the combined targeted inhibition of STAT1, ARTD8, ARTD9 and/or DTX3L could increase the efficacy of chemotherapy or radiation treatment in prostate and other high-risk tumor types with an increased STAT1 signaling.


Subject(s)
Interferon Regulatory Factor-1/genetics , Neoplasm Proteins/genetics , Poly(ADP-ribose) Polymerases/genetics , Prostatic Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Cell Movement/genetics , Cell Proliferation , Humans , Interferon Regulatory Factor-1/metabolism , Male , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/pathology , STAT1 Transcription Factor/genetics , Ubiquitin-Protein Ligases/metabolism
4.
BMC Vet Res ; 9: 141, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23870652

ABSTRACT

BACKGROUND: The TSH stimulation test to confirm canine hypothyroidism is commonly performed using a recombinant human TSH (rhTSH), as up to date, canine TSH is not yet commercially available. Limiting factors for the use of rhTSH are its high costs and occasional difficulties in product availability. Less expensive bovine TSH preparations (bTSH) purified from bovine pituitary glands are readily commercially available. The aim of this study was to evaluate two different bTSH products as alternative to rhTSH using mass spectrometry. RESULTS: More than 50 proteins, including other pituitary hormones, bovine albumin, hemoglobin, and tissue proteins were identified in the bTSH preparations. In contrast, rhTSH proved to be a highly pure product. Significantly higher endotoxin levels could be detected in all bTSH products compared to the rhTSH. CONCLUSIONS: Both bTSH products are crude mixtures and therefore not an acceptable alternative to rhTSH. Their use should be discouraged to prevent unintended side effects.


Subject(s)
Endotoxins/analysis , Mass Spectrometry/veterinary , Recombinant Proteins/analysis , Thyrotropin/analysis , Animals , Cattle , Electrophoresis, Polyacrylamide Gel/veterinary , Enzyme-Linked Immunosorbent Assay/veterinary , Growth Hormone/analysis , Humans , Luteinizing Hormone/analysis , Tissue Extracts/chemistry
5.
Nat Struct Mol Biol ; 20(4): 502-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23474714

ABSTRACT

ADP-ribosylation is an important post-translational protein modification (PTM) that regulates diverse biological processes. ADP-ribosyltransferase diphtheria toxin-like 10 (ARTD10, also known as PARP10) mono-ADP-ribosylates acidic side chains and is one of eighteen ADP-ribosyltransferases that catalyze mono- or poly-ADP-ribosylation of target proteins. Currently, no enzyme is known that reverses ARTD10-catalyzed mono-ADP-ribosylation. Here we report that ARTD10-modified targets are substrates for the macrodomain proteins MacroD1, MacroD2 and C6orf130 from Homo sapiens as well as for the macrodomain protein Af1521 from archaebacteria. Structural modeling and mutagenesis of MacroD1 and MacroD2 revealed a common core structure with Asp102 and His106 of MacroD2 implicated in the hydrolytic reaction. Notably, MacroD2 reversed the ARTD10-catalyzed, mono-ADP-ribose-mediated inhibition of glycogen synthase kinase 3ß (GSK3ß) in vitro and in cells, thus underlining the physiological and regulatory importance of mono-ADP-ribosylhydrolase activity. Our results establish macrodomain-containing proteins as mono-ADP-ribosylhydrolases and define a class of enzymes that renders mono-ADP-ribosylation a reversible modification.


Subject(s)
N-Glycosyl Hydrolases/metabolism , Adenosine Diphosphate Ribose/metabolism , Humans , Models, Molecular , Mutagenesis , N-Glycosyl Hydrolases/chemistry , N-Glycosyl Hydrolases/genetics
6.
J Cell Sci ; 126(Pt 9): 1969-80, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23487038

ABSTRACT

The B-aggressive lymphoma-1 protein and ADP-ribosyltransferase BAL1/ARTD9 has been recently identified as a risk-related gene product in aggressive diffuse large B-cell lymphoma (DLBCL). BAL1 is constitutively expressed in a subset of high-risk DLBCLs with an active host inflammatory response and has been suggested to be associated with interferon-related gene expression. Here we identify BAL1 as a novel oncogenic survival factor in DLBCL and show that constitutive overexpression of BAL1 in DLBCL tightly associates with intrinsic interferon-gamma (IFNγ) signaling and constitutive activity of signal transducer and activator of transcription (STAT)-1. Remarkably, BAL1 stimulates the phosphorylation of both STAT1 isoforms, STAT1α and STAT1ß, on Y701 and thereby promotes the nuclear accumulation of the antagonistically acting and transcriptionally repressive isoform STAT1ß. Moreover, BAL1 physically interacts with both STAT1α and STAT1ß through its macrodomains in an ADP-ribosylation-dependent manner. BAL1 directly inhibits, together with STAT1ß, the expression of tumor suppressor and interferon response factor (IRF)-1. Conversely, BAL1 enhances the expression of the proto-oncogenes IRF2 and B-cell CLL/lymphoma (BCL)-6 in DLBCL. Our results show for the first time that BAL1 represses the anti-proliferative and pro-apoptotic IFNγ-STAT1-IRF1-p53 axis and mediates proliferation, survival and chemo-resistance in DLBCL. As a consequence constitutive IFNγ-STAT1 signaling does not lead to apoptosis but rather to chemo-resistance in DLBCL overexpressing BAL1. Our results suggest that BAL1 may induce an switch in STAT1 from a tumor suppressor to an oncogene in high-risk DLBCL.


Subject(s)
Apoptosis , Cell Proliferation , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Neoplasm Proteins/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-2/biosynthesis , Interferon Regulatory Factor-2/genetics , Interferon-gamma/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Neoplasm Proteins/genetics , Poly(ADP-ribose) Polymerases , Protein Isoforms/genetics , Protein Isoforms/metabolism , STAT1 Transcription Factor/genetics , Tumor Suppressor Protein p53/genetics
7.
Nat Struct Mol Biol ; 19(7): 685-692, 2012 Jun 10.
Article in English | MEDLINE | ID: mdl-22683995

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP1) is a primary DNA damage sensor whose (ADP-ribose) polymerase activity is acutely regulated by interaction with DNA breaks. Upon activation at sites of DNA damage, PARP1 modifies itself and other proteins by covalent addition of long, branched polymers of ADP-ribose, which in turn recruit downstream DNA repair and chromatin remodeling factors. PARP1 recognizes DNA damage through its N-terminal DNA-binding domain (DBD), which consists of a tandem repeat of an unusual zinc-finger (ZnF) domain. We have determined the crystal structure of the human PARP1-DBD bound to a DNA break. Along with functional analysis of PARP1 recruitment to sites of DNA damage in vivo, the structure reveals a dimeric assembly whereby ZnF1 and ZnF2 domains from separate PARP1 molecules form a strand-break recognition module that helps activate PARP1 by facilitating its dimerization and consequent trans-automodification.


Subject(s)
DNA Damage , Poly(ADP-ribose) Polymerases/metabolism , Zinc Fingers , DNA/metabolism , Dimerization , Models, Molecular , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/chemistry
8.
Mol Cell ; 46(2): 200-11, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22464733

ABSTRACT

Caspase 1 is part of the inflammasome, which is assembled upon pathogen recognition, while caspases 3 and/or 7 are mediators of apoptotic and nonapoptotic functions. PARP1 cleavage is a hallmark of apoptosis yet not essential, suggesting it has another physiological role. Here we show that after LPS stimulation, caspase 7 is activated by caspase 1, translocates to the nucleus, and cleaves PARP1 at the promoters of a subset of NF-κB target genes negatively regulated by PARP1. Mutating the PARP1 cleavage site D214 renders PARP1 uncleavable and inhibits PARP1 release from chromatin and chromatin decondensation, thereby restraining the expression of cleavage-dependent NF-κB target genes. These findings propose an apoptosis-independent regulatory role for caspase 7-mediated PARP1 cleavage in proinflammatory gene expression and provide insight into inflammasome signaling.


Subject(s)
Caspase 7/physiology , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/physiology , Chromatin/metabolism , Gene Expression Regulation , Humans , Inflammation/genetics , Mice , Mutation , NLR Family, Pyrin Domain-Containing 3 Protein , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Signal Transduction
9.
Biochem Biophys Res Commun ; 414(4): 641-6, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21982765

ABSTRACT

BACKGROUND: Generation of reactive oxygen species (ROS) is a key feature of vascular disease. Activation of the nuclear enzyme poly (adenosine diphosphate [ADP]-ribose) polymerase-1 (PARP-1) is a downstream effector of oxidative stress. METHODS: PARP-1(-/-) and PARP-1(+/+) mice were injected with paraquat (PQ; 10 mg/kg i.p.) to induce intracellular oxidative stress. Aortic rings were suspended in organ chambers for isometric tension recording to analyze vascular function. RESULTS: PQ treatment markedly impaired endothelium-dependent relaxations to acetylcholine in PARP-1(-/-), but not PARP-1(+/+) mice (p<0.0001). Maximal relaxation was 45% in PQ treated PARP-1(-/-) mice compared to 79% in PARP-1(+/+) mice. In contrast, endothelium-independent relaxations to sodium nitroprusside (SNP) were not altered. After PQ treatment, l-NAME enhanced contractions to norepinephrine by 2.0-fold in PARP-1(-/-) mice, and those to acetylcholine by 3.3-fold, respectively, as compared to PARP-1(+/+) mice. PEG-superoxide dismutase (SOD) and PEG-catalase prevented the effect of PQ on endothelium-dependent relaxations to acetylcholine in PARP-1(-/-) mice (p<0.001 vs. PQ treated PARP-1(+/+) mice. Indomethacin restored endothelium-dependent relaxations to acetylcholine in PQ treated PARP-1(-/-) mice (p<0.05 vs. PQ treated PARP-1(+/+). CONCLUSION: PARP-1 protects from acute intracellular oxidative stress induced endothelial dysfunction by inhibiting ROS induced production of vasoconstrictor prostanoids.


Subject(s)
Endothelium, Vascular/physiology , Oxidative Stress/physiology , Poly(ADP-ribose) Polymerases/physiology , Vasoconstriction/physiology , Vasodilation/physiology , Animals , Endothelium, Vascular/drug effects , Mice , Mice, Mutant Strains , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Oxidative Stress/genetics , Paraquat/pharmacology , Peroxynitrous Acid/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/biosynthesis , Reactive Oxygen Species/metabolism , Vasoconstriction/genetics , Vasodilation/genetics
10.
Trends Biochem Sci ; 35(4): 208-19, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20106667

ABSTRACT

ADP-ribosylation is a post-translational modification of proteins catalyzed by ADP-ribosyltransferases. It comprises the transfer of the ADP-ribose moiety from NAD+ to specific amino acid residues on substrate proteins or to ADP-ribose itself. Currently, 22 human genes encoding proteins that possess an ADP-ribosyltransferase catalytic domain are known. Recent structural and enzymological evidence of poly(ADP-ribose)polymerase (PARP) family members demonstrate that earlier proposed names and classifications of these proteins are no longer accurate. Here we summarize these new findings and propose a new consensus nomenclature for all ADP-ribosyltransferases (ARTs) based on the catalyzed reaction and on structural features. A unified nomenclature would facilitate communication between researchers both inside and outside the ADP-ribosylation field.


Subject(s)
ADP Ribose Transferases/classification , ADP Ribose Transferases/metabolism , Mammals , Terminology as Topic , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Animals , Catalytic Domain/genetics , Catalytic Domain/physiology , Humans , Mammals/genetics , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/classification , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational
11.
Nat Struct Mol Biol ; 16(9): 923-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19680243

ABSTRACT

Poly-ADP-ribosylation is a post-translational modification catalyzed by PARP enzymes with roles in transcription and chromatin biology. Here we show that distinct macrodomains, including those of histone macroH2A1.1, are recruited to sites of PARP1 activation induced by laser-generated DNA damage. Chemical PARP1 inhibitors, PARP1 knockdown and mutation of ADP-ribose-binding residues in macroH2A1.1 abrogate macrodomain recruitment. Notably, histone macroH2A1.1 senses PARP1 activation, transiently compacts chromatin, reduces the recruitment of DNA damage factor Ku70-Ku80 and alters gamma-H2AX patterns, whereas the splice variant macroH2A1.2, which is deficient in poly-ADP-ribose binding, does not mediate chromatin rearrangements upon PARP1 activation. The structure of the macroH2A1.1 macrodomain in complex with ADP-ribose establishes a poly-ADP-ribose cap-binding function and reveals conformational changes in the macrodomain upon ligand binding. We thus identify macrodomains as modules that directly sense PARP activation in vivo and establish macroH2A histones as dynamic regulators of chromatin plasticity.


Subject(s)
Chromatin , Histones/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Amino Acid Motifs , DNA Damage , Enzyme Activation , HeLa Cells , Histones/chemistry , Humans , Models, Molecular , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/chemistry , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , RNA Processing, Post-Transcriptional
12.
Nucleic Acids Res ; 37(11): 3723-38, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19372272

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP1) synthesizes poly(ADP-ribose) (PAR) using nicotinamide adenine dinucleotide (NAD) as a substrate. Despite intensive research on the cellular functions of PARP1, the molecular mechanism of PAR formation has not been comprehensively understood. In this study, we elucidate the molecular mechanisms of poly(ADP-ribosyl)ation and identify PAR acceptor sites. Generation of different chimera proteins revealed that the amino-terminal domains of PARP1, PARP2 and PARP3 cooperate tightly with their corresponding catalytic domains. The DNA-dependent interaction between the amino-terminal DNA-binding domain and the catalytic domain of PARP1 increased V(max) and decreased the K(m) for NAD. Furthermore, we show that glutamic acid residues in the auto-modification domain of PARP1 are not required for PAR formation. Instead, we identify individual lysine residues as acceptor sites for ADP-ribosylation. Together, our findings provide novel mechanistic insights into PAR synthesis with significant relevance for the different biological functions of PARP family members.


Subject(s)
Lysine/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/chemistry , Catalytic Domain , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , DNA/metabolism , Glutamic Acid/metabolism , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Multimerization , Protein Structure, Tertiary
13.
Science ; 324(5926): 513-6, 2009 Apr 24.
Article in English | MEDLINE | ID: mdl-19390046

ABSTRACT

Polyphosphate (polyP) occurs ubiquitously in cells, but its functions are poorly understood and its synthesis has only been characterized in bacteria. Using x-ray crystallography, we identified a eukaryotic polyphosphate polymerase within the membrane-integral vacuolar transporter chaperone (VTC) complex. A 2.6 angstrom crystal structure of the catalytic domain grown in the presence of adenosine triphosphate (ATP) reveals polyP winding through a tunnel-shaped pocket. Nucleotide- and phosphate-bound structures suggest that the enzyme functions by metal-assisted cleavage of the ATP gamma-phosphate, which is then in-line transferred to an acceptor phosphate to form polyP chains. Mutational analysis of the transmembrane domain indicates that VTC may integrate cytoplasmic polymer synthesis with polyP membrane translocation. Identification of the polyP-synthesizing enzyme opens the way to determine the functions of polyP in lower eukaryotes.


Subject(s)
Membrane Proteins/chemistry , Phosphotransferases/chemistry , Polyphosphates/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Biological Transport , Catalysis , Catalytic Domain , Crystallography, X-Ray , Membrane Proteins/metabolism , Models, Molecular , Phosphotransferases/metabolism , Polyphosphates/metabolism , Protein Conformation , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
14.
Front Biosci (Landmark Ed) ; 14(1): 72-111, 2009 01 01.
Article in English | MEDLINE | ID: mdl-19273055

ABSTRACT

The current literature clearly indicates that PARP1 but also PARP2 play a pivotal role in modulating the cellular responses to stress. Genetic and pharmacological studies demonstrated that overactivation of PARP1 is a key mediator of programmed-necrotic cell death in vivo. PARP1 appears to be also involved in programmed cell death processes others than necrosis, such as apoptosis or macroautophagocytotic cell death. On the other hand, growing evidence suggests that both PARP1 and PARP2 are multi-faced enzymes also playing important roles in cell survival processes. PARP1 and PARP2 were shown to be required for the maintenance of genomic integrity and to act as a survival factor for highly proliferating cells such as stem cells but also non-proliferating neuronal cells against cell death induced by oxidative stress under mild and moderate progressive damage in vivo. This review briefly summarizes the recent findings, which support a crucial role of PARP1 in different programmed cell death and cell survival processes. A special focus is placed on the proposed molecular mechanisms underlying the "Jekyll and Hyde" duality of PARP1 in cell death and cell survival pathways. A potential crosstalk between PARP1, PARP2 and other NAD+-dependent ADP-ribosyling enzymes such as Sirtuins and CD38 in cell death and survival pathways is discussed.


Subject(s)
Cell Death/physiology , Cell Survival/physiology , Poly(ADP-ribose) Polymerases/physiology , Animals , Humans , Poly (ADP-Ribose) Polymerase-1
15.
Mol Cell ; 32(1): 57-69, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18851833

ABSTRACT

ADP-ribosylation controls many processes, including transcription, DNA repair, and bacterial toxicity. ADP-ribosyltransferases and poly-ADP-ribose polymerases (PARPs) catalyze mono- and poly-ADP-ribosylation, respectively, and depend on a highly conserved glutamate residue in the active center for catalysis. However, there is an apparent absence of this glutamate for the recently described PARP6-PARP16, raising questions about how these enzymes function. We find that PARP10, in contrast to PARP1, lacks the catalytic glutamate and has transferase rather than polymerase activity. Despite this fundamental difference, PARP10 also modifies acidic residues. Consequently, we propose an alternative catalytic mechanism for PARP10 compared to PARP1 in which the acidic target residue of the substrate functionally substitutes for the catalytic glutamate by using substrate-assisted catalysis to transfer ADP-ribose. This mechanism explains why the novel PARPs are unable to function as polymerases. This discovery will help to illuminate the different biological functions of mono- versus poly-ADP-ribosylation in cells.


Subject(s)
ADP Ribose Transferases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/metabolism , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Amino Acid Sequence , Catalytic Domain , Cell Line , Conserved Sequence , Humans , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
16.
BMC Cell Biol ; 9: 39, 2008 Jul 21.
Article in English | MEDLINE | ID: mdl-18644123

ABSTRACT

BACKGROUND: The enzymes responsible for the synthesis of poly-ADP-ribose are named poly-ADP-ribose polymerases (PARP). PARP-2 is a nuclear protein, which regulates a variety of cellular functions that are mainly controlled by protein-protein interactions. A previously described non-conventional bipartite nuclear localization sequence (NLS) lies in the amino-terminal DNA binding domain of PARP-2 between amino acids 1-69; however, this targeting sequence has not been experimentally examined or validated. RESULTS: Using a site-directed mutagenesis approach, we found that lysines 19 and 20, located within a previously described bipartite NLS, are not required for nuclear localization of PARP-2. In contrast, lysine 36, which is located within a predicted classical monopartite NLS, was required for PARP-2 nuclear localization. While wild type PARP-2 interacted with importin alpha3 and to a very weak extent with importin alpha1 and importin alpha5, the mutant PARP-2 (K36R) did not interact with importin alpha3, providing a molecular explanation why PARP-2 (K36R) is not targeted to the nucleus. CONCLUSION: Our results provide strong evidence that lysine 36 of PARP-2 is a critical residue for proper nuclear targeting of PARP-2 and consequently for the execution of its biological functions.


Subject(s)
Cell Nucleus/metabolism , Lysine/metabolism , Nuclear Localization Signals , Poly(ADP-ribose) Polymerases , alpha Karyopherins/metabolism , Amino Acid Sequence , Animals , Antibiotics, Antineoplastic/metabolism , Cell Line , Fatty Acids, Unsaturated/metabolism , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , alpha Karyopherins/genetics
17.
Int J Biochem Cell Biol ; 40(10): 2274-83, 2008.
Article in English | MEDLINE | ID: mdl-18436469

ABSTRACT

Poly-ADP-ribose polymerase-2 (PARP-2) was described to regulate cellular functions comprising DNA surveillance, inflammation and cell differentiation by co-regulating different transcription factors. Using an in vitro and in vivo approach, we identified PARP-2 as a new substrate for the histone acetyltransferases PCAF and GCN5L. Site directed mutagenesis indicated that lysines 36 and 37, located in the nuclear localization signal of PARP-2, are the main targets for PCAF and GCN5L activity in vitro. Interestingly, acetylation of the same two PARP-2 residues reduces the DNA binding and enzymatic activity of PARP-2. Finally, PARP-2 with mutated lysines 36 and 37 showed reduced auto-mono-ADP-ribosylation when compared to wild type PARP-2. Together, our results provide evidence that acetylation of PARP-2 is a key post-translational modification that may regulate DNA binding and consequently also the enzymatic activity of PARP-2.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Lysine/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Acetylation , Amino Acid Sequence , Animals , Cell Line , DNA/metabolism , Humans , Mice , Molecular Sequence Data , Mutant Proteins/metabolism , Nerve Tissue Proteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/chemistry , Protein Binding , p300-CBP Transcription Factors/metabolism
18.
Mol Cancer Res ; 6(2): 282-90, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18314489

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is the key transcription factor regulating hypoxia-dependent gene expression. Lack of oxygen stabilizes HIF-1, which in turn modulates the gene expression pattern to adapt cells to the hypoxic environment. Activation of HIF-1 is also detected in most solid tumors and supports tumor growth through the expression of target genes that are involved in processes like cell proliferation, energy metabolism, and oxygen delivery. Poly(ADP-ribose) polymerase 1 (PARP1) is a chromatin-associated protein, which was shown to regulate transcription. Here we report that chronic myelogenous leukemia cells expressing small interfering RNA against PARP1, which were injected into wild-type mice expressing PARP1, showed tumor growth with increased levels of necrosis, limited vascularization, and reduced expression of GLUT-1. Of note, PARP1-deficient cells showed a reduced HIF-1 transcriptional activation that was dependent on PARP1 enzymatic activity. PARP1 neither influenced binding of HIF-1 to its hypoxic response element nor changed HIF-1alpha protein levels in hypoxic cells. However, PARP1 formed a complex with HIF-1alpha through direct protein interaction and coactivated HIF-1alpha-dependent gene expression. These findings provide convincing evidence that wild-type mice expressing PARP1 cannot compensate for the loss of PARP1 in tumor cells and strengthen the importance of the role of PARP1 as a transcriptional coactivator of HIF-1-dependent gene expression during tumor progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1/metabolism , Neoplasms/genetics , Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Cell Death , Cell Proliferation , Cell Survival , Down-Regulation/genetics , Fibroblasts/enzymology , Fibroblasts/metabolism , HeLa Cells , Humans , K562 Cells , Lung/cytology , Lung/enzymology , Mice , Mice, Nude , Necrosis , Neoplasms/blood supply , Poly(ADP-ribose) Polymerases/deficiency , Poly(ADP-ribose) Polymerases/genetics , Protein Binding , RNA, Small Interfering , Signal Transduction , Transcription, Genetic , Transcriptional Activation/genetics
19.
J Mol Biol ; 377(3): 668-78, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18280497

ABSTRACT

Nuclear factor kappa B (NF-kappaB) plays an important role in the transcriptional regulation of genes involved in inflammation and cell survival. Transcriptional coactivators that methylate histones become increasingly important. Recently, we provided evidence that coactivator-associated arginine methyltransferase 1 (CARM1) is a transcriptional coactivator of NF-kappaB and functions as a promoter-specific regulator of NF-kappaB recruitment to chromatin. Here, we show that protein arginine methyltransferase 1 (PRMT1) synergistically coactivates NF-kappaB-dependent gene expression at the macrophage inflammatory protein 2 and human immunodeficiency virus 1 long terminal repeat promoters in concert with the transcriptional coactivators p300/CREB binding protein, CARM1, and poly(ADP-ribose) polymerase 1. PRMT1 formed a complex with poly(ADP-ribose) polymerase 1 and NF-kappaB in vivo and interacted directly with the NF-kappaB subunit p65 in vitro. The methyltransferase activity of PRMT1 appeared essential for its coactivator function in context with CARM1 and p300/CREB binding protein. These results suggest that the cooperative action between PRMT1 and CARM1 is required for NF-kappaB-dependent gene expression.


Subject(s)
NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/physiology , Protein-Arginine N-Methyltransferases/physiology , Trans-Activators/physiology , Animals , CREB-Binding Protein/physiology , Cells, Cultured , Chemokine CXCL2/genetics , Gene Expression Regulation , HIV Long Terminal Repeat/genetics , Humans , Mice , Mice, Knockout , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/metabolism , Transcription Factor RelA/metabolism
20.
Front Biosci ; 13: 3046-82, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17981777

ABSTRACT

Poly-ADP-ribose metabolism plays a mayor role in a wide range of biological processes, such as maintenance of genomic stability, transcriptional regulation, energy metabolism and cell death. Poly-ADP-ribose polymerases (PARPs) are an ancient family of enzymes, as evidenced by the poly-ADP-ribosylating activities reported in dinoflagellates and archaebacteria and by the identification of Parp-like genes in eubacterial and archaeabacterial genomes. Six genes encoding "bona fide" PARP enzymes have been identified in mammalians: PARP1, PARP2, PARP3, PARP4/vPARP, PARP5/Tankyrases-1 and PARP6/Tankyrases-2. The best studied of these enzymes PARP1 plays a primary role in the process of poly-ADP-ribosylation. PARP1-mediated poly-ADP-ribosylation has been implicated in the pathogenesis of cancer, inflammatory and neurodegenerative disorders. This review will summarize the novel findings and concepts for PARP enzymes and their poly-ADP-ribosylation activity in the regulation of physiological and pathophysiological processes. A special focus is placed on the proposed molecular mechanisms involved in these processes, such as signaling, regulation of telomere dynamics, remodeling of chromatin structure and transcriptional regulation. A potential functional cross talk between PARP family members and other NAD+-consuming enzymes is discussed.


Subject(s)
Poly(ADP-ribose) Polymerases/physiology , Adenosine Diphosphate/chemistry , Animals , Cell Death , Chromatin/chemistry , Chromatin Immunoprecipitation , Crystallization , Humans , Mass Spectrometry/methods , Mice , Mice, Transgenic , Multigene Family , Poly(ADP-ribose) Polymerases/chemistry , Protein Binding , Protein Isoforms , Ribose/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL