Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(16)2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37628794

ABSTRACT

Our understanding of the molecular mechanisms underlying cancer development and evolution have evolved rapidly over recent years, and the variation from one patient to another is now widely recognized. Consequently, one-size-fits-all approaches to the treatment of cancer have been superseded by precision medicines that target specific disease characteristics, promising maximum clinical efficacy, minimal safety concerns, and reduced economic burden. While precision oncology has been very successful in the treatment of some tumors with specific characteristics, a large number of patients do not yet have access to precision medicines for their disease. The success of next-generation precision oncology depends on the discovery of new actionable disease characteristics, rapid, accurate, and comprehensive diagnosis of complex phenotypes within each patient, novel clinical trial designs with improved response rates, and worldwide access to novel targeted anticancer therapies for all patients. This review outlines some of the current technological trends, and highlights some of the complex multidisciplinary efforts that are underway to ensure that many more patients with cancer will be able to benefit from precision oncology in the near future.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Precision Medicine , Medical Oncology , Interdisciplinary Studies , Phenotype
2.
DNA Repair (Amst) ; 58: 52-61, 2017 10.
Article in English | MEDLINE | ID: mdl-28866241

ABSTRACT

Acylpeptide hydrolase (APEH) deacetylates N-alpha-acetylated peptides and selectively degrades oxidised proteins, but the biochemical pathways that are regulated by this protease are unknown. Here, we identify APEH as a component of the cellular response to DNA damage. Although APEH is primarily localised in the cytoplasm, we show that a sub-fraction of this enzyme is sequestered at sites of nuclear damage following UVA irradiation or following oxidative stress. We show that localization of APEH at sites of nuclear damage is mediated by direct interaction with XRCC1, a scaffold protein that accelerates the repair of DNA single-strand breaks. We show that APEH interacts with the amino-terminal domain of XRCC1, and that APEH facilitates both single-strand break repair and cell survival following exposure to H2O2 in human cells. These data identify APEH as a novel proteolytic component of the DNA damage response.


Subject(s)
DNA Breaks, Single-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Peptide Hydrolases/metabolism , DNA/drug effects , DNA/metabolism , Humans , Hydrogen Peroxide/toxicity , Protein Binding , Protein Interaction Domains and Motifs , X-ray Repair Cross Complementing Protein 1
3.
Bioessays ; 39(3)2017 03.
Article in English | MEDLINE | ID: mdl-28133776

ABSTRACT

Non-homologous end-joining (NHEJ) is the dominant means of repairing chromosomal DNA double strand breaks (DSBs), and is essential in human cells. Fifteen or more proteins can be involved in the detection, signalling, synapsis, end-processing and ligation events required to repair a DSB, and must be assembled in the confined space around the DNA ends. We review here a number of interaction points between the core NHEJ components (Ku70, Ku80, DNA-PKcs, XRCC4 and Ligase IV) and accessory factors such as kinases, phosphatases, polymerases and structural proteins. Conserved protein-protein interaction sites such as Ku-binding motifs (KBMs), XLF-like motifs (XLMs), FHA and BRCT domains illustrate that different proteins compete for the same binding sites on the core machinery, and must be spatially and temporally regulated. We discuss how post-translational modifications such as phosphorylation, ADP-ribosylation and ubiquitinylation may regulate sequential steps in the NHEJ pathway or control repair at different types of DNA breaks.


Subject(s)
DNA End-Joining Repair , Amino Acid Sequence , Animals , DNA Breaks, Double-Stranded , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/physiology , Humans , Protein Interaction Domains and Motifs , Protein Interaction Maps
4.
Nature ; 541(7635): 87-91, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28002403

ABSTRACT

XRCC1 is a molecular scaffold protein that assembles multi-protein complexes involved in DNA single-strand break repair. Here we show that biallelic mutations in the human XRCC1 gene are associated with ocular motor apraxia, axonal neuropathy, and progressive cerebellar ataxia. Cells from a patient with mutations in XRCC1 exhibited not only reduced rates of single-strand break repair but also elevated levels of protein ADP-ribosylation. This latter phenotype is recapitulated in a related syndrome caused by mutations in the XRCC1 partner protein PNKP and implicates hyperactivation of poly(ADP-ribose) polymerase/s as a cause of cerebellar ataxia. Indeed, remarkably, genetic deletion of Parp1 rescued normal cerebellar ADP-ribose levels and reduced the loss of cerebellar neurons and ataxia in Xrcc1-defective mice, identifying a molecular mechanism by which endogenous single-strand breaks trigger neuropathology. Collectively, these data establish the importance of XRCC1 protein complexes for normal neurological function and identify PARP1 as a therapeutic target in DNA strand break repair-defective disease.


Subject(s)
Cerebellar Ataxia/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mutation , Poly (ADP-Ribose) Polymerase-1/metabolism , Adenosine Diphosphate Ribose/metabolism , Alleles , Animals , Apraxias/congenital , Apraxias/genetics , Ataxia/genetics , Axons/pathology , Cerebellar Ataxia/pathology , Cerebellum/metabolism , Cerebellum/pathology , Chromatin/metabolism , Cogan Syndrome/genetics , DNA Breaks, Single-Stranded , DNA Repair/genetics , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/deficiency , Female , Humans , Interneurons/metabolism , Interneurons/pathology , Male , Mice , Pedigree , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Poly (ADP-Ribose) Polymerase-1/deficiency , Poly (ADP-Ribose) Polymerase-1/genetics , X-ray Repair Cross Complementing Protein 1
5.
Cancer Res ; 76(20): 6084-6094, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27550455

ABSTRACT

The PARP inhibitor AZD2461 was developed as a next-generation agent following olaparib, the first PARP inhibitor approved for cancer therapy. In BRCA1-deficient mouse models, olaparib resistance predominantly involves overexpression of P-glycoprotein, so AZD2461 was developed as a poor substrate for drug transporters. Here we demonstrate the efficacy of this compound against olaparib-resistant tumors that overexpress P-glycoprotein. In addition, AZD2461 was better tolerated in combination with chemotherapy than olaparib in mice, which suggests that AZD2461 could have significant advantages over olaparib in the clinic. However, this superior toxicity profile did not extend to rats. Investigations of this difference revealed a differential PARP3 inhibitory activity for each compound and a higher level of PARP3 expression in bone marrow cells from mice as compared with rats and humans. Our findings have implications for the use of mouse models to assess bone marrow toxicity for DNA-damaging agents and inhibitors of the DNA damage response. Finally, structural modeling of the PARP3-active site with different PARP inhibitors also highlights the potential to develop compounds with different PARP family member specificity profiles for optimal antitumor activity and tolerability. Cancer Res; 76(20); 6084-94. ©2016 AACR.


Subject(s)
Neoplasms, Experimental/drug therapy , Phthalazines/pharmacology , Piperidines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Animals , Bone Marrow/drug effects , Cell Line, Tumor , DNA Damage , DNA Repair , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Discovery , Genes, BRCA1 , Humans , Mice , Phthalazines/administration & dosage , Phthalazines/toxicity , Piperazines/administration & dosage , Piperidines/toxicity , Poly(ADP-ribose) Polymerases/chemistry , Rats , Temozolomide , Xenograft Model Antitumor Assays
6.
Nat Commun ; 7: 12404, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27530147

ABSTRACT

PARP3 is a member of the ADP-ribosyl transferase superfamily that we show accelerates the repair of chromosomal DNA single-strand breaks in avian DT40 cells. Two-dimensional nuclear magnetic resonance experiments reveal that PARP3 employs a conserved DNA-binding interface to detect and stably bind DNA breaks and to accumulate at sites of chromosome damage. PARP3 preferentially binds to and is activated by mononucleosomes containing nicked DNA and which target PARP3 trans-ribosylation activity to a single-histone substrate. Although nicks in naked DNA stimulate PARP3 autoribosylation, nicks in mononucleosomes promote the trans-ribosylation of histone H2B specifically at Glu2. These data identify PARP3 as a molecular sensor of nicked nucleosomes and demonstrate, for the first time, the ribosylation of chromatin at a site-specific DNA single-strand break.


Subject(s)
DNA Breaks, Single-Stranded , Histones/metabolism , Nucleosomes/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Ribose/metabolism , Animals , Cell Line , Chickens , Chromatin/metabolism , Chromosomes/metabolism , DNA/metabolism , DNA Repair , Humans , Models, Molecular , Poly(ADP-ribose) Polymerases/chemistry , Protein Domains
7.
Nat Commun ; 7: 11242, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27063109

ABSTRACT

The Ku-binding motif (KBM) is a short peptide module first identified in APLF that we now show is also present in Werner syndrome protein (WRN) and in Modulator of retrovirus infection homologue (MRI). We also identify a related but functionally distinct motif in XLF, WRN, MRI and PAXX, which we denote the XLF-like motif. We show that WRN possesses two KBMs; one at the N terminus next to the exonuclease domain and one at the C terminus next to an XLF-like motif. We reveal that the WRN C-terminal KBM and XLF-like motif function cooperatively to bind Ku complexes and that the N-terminal KBM mediates Ku-dependent stimulation of WRN exonuclease activity. We also show that WRN accelerates DSB repair by a mechanism requiring both KBMs, demonstrating the importance of WRN interaction with Ku. These data define a conserved family of KBMs that function as molecular tethers to recruit and/or stimulate enzymes during NHEJ.


Subject(s)
Antigens, Nuclear/metabolism , Conserved Sequence , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , DNA Breaks, Double-Stranded , DNA Damage , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Humans , Ku Autoantigen , Models, Biological , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RecQ Helicases/chemistry , RecQ Helicases/metabolism , Werner Syndrome Helicase
8.
Front Genet ; 7: 52, 2016.
Article in English | MEDLINE | ID: mdl-27092175

ABSTRACT

Impulsivity is associated with a spectrum of psychiatric disorders including drug addiction. To investigate genetic associations with impulsivity and initiation of drug taking, we took a two-step approach. First, we identified genes whose expression level in prefrontal cortex, striatum and accumbens were associated with impulsive behavior in the 5-choice serial reaction time task across 10 BXD recombinant inbred (BXD RI) mouse strains and their progenitor C57BL/6J and DBA2/J strains. Behavioral data were correlated with regional gene expression using GeneNetwork (www.genenetwork.org), to identify 44 genes whose probability of association with impulsivity exceeded a false discovery rate of < 0.05. We then interrogated the IMAGEN database of 1423 adolescents for potential associations of SNPs in human homologs of those genes identified in the mouse study, with brain activation during impulsive performance in the Monetary Incentive Delay task, and with novelty seeking scores from the Temperament and Character Inventory, as well as alcohol experience. There was a significant overall association between the human homologs of impulsivity-related genes and percentage of premature responses in the MID task and with fMRI BOLD-response in ventral striatum (VS) during reward anticipation. In contrast, no significant association was found between the polygenic scores and anterior cingulate cortex activation. Univariate association analyses revealed that the G allele (major) of the intronic SNP rs6438839 in the KALRN gene was significantly associated with increased VS activation. Additionally, the A-allele (minor) of KALRN intronic SNP rs4634050, belonging to the same haplotype block, was associated with increased frequency of binge drinking.

9.
DNA Repair (Amst) ; 35: 116-25, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26519825

ABSTRACT

Aprataxin, aprataxin and PNKP-like factor (APLF) and polynucleotide kinase phosphatase (PNKP) are key DNA-repair proteins with diverse functions but which all contain a homologous forkhead-associated (FHA) domain. Their primary binding targets are casein kinase 2-phosphorylated forms of the XRCC1 and XRCC4 scaffold molecules which respectively coordinate single-stranded and double-stranded DNA break repair pathways. Here, we present the high-resolution X-ray structure of a complex of phosphorylated XRCC4 with APLF, the most divergent of the three FHA domain family members. This, combined with NMR and biochemical analysis of aprataxin and APLF binding to singly and multiply-phosphorylated forms of XRCC1 and XRCC4, and comparison with PNKP reveals a pattern of distinct but overlapping binding specificities that are differentially modulated by multi-site phosphorylation. Together, our data illuminate important differences between activities of the three phospho-binding domains, in spite of a close evolutionary relationship between them.


Subject(s)
DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-Binding Proteins/chemistry , Nuclear Proteins/chemistry , Amino Acid Sequence , Binding Sites , Casein Kinase II/metabolism , Crystallography, X-Ray , DNA Repair , DNA Repair Enzymes/ultrastructure , DNA-(Apurinic or Apyrimidinic Site) Lyase/ultrastructure , DNA-Binding Proteins/ultrastructure , Humans , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/ultrastructure , Poly-ADP-Ribose Binding Proteins , Protein Structure, Tertiary , X-ray Repair Cross Complementing Protein 1
10.
Nucleic Acids Res ; 43(14): 6934-44, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26130715

ABSTRACT

Poly (ADP-ribose) is synthesized at DNA single-strand breaks and can promote the recruitment of the scaffold protein, XRCC1. However, the mechanism and importance of this process has been challenged. To address this issue, we have characterized the mechanism of poly (ADP-ribose) binding by XRCC1 and examined its importance for XRCC1 function. We show that the phosphate-binding pocket in the central BRCT1 domain of XRCC1 is required for selective binding to poly (ADP-ribose) at low levels of ADP-ribosylation, and promotes interaction with cellular PARP1. We also show that the phosphate-binding pocket is required for EGFP-XRCC1 accumulation at DNA damage induced by UVA laser, H2O2, and at sites of sub-nuclear PCNA foci, suggesting that poly (ADP-ribose) promotes XRCC1 recruitment both at single-strand breaks globally across the genome and at sites of DNA replication stress. Finally, we show that the phosphate-binding pocket is required following DNA damage for XRCC1-dependent acceleration of DNA single-strand break repair, DNA base excision repair, and cell survival. These data support the hypothesis that poly (ADP-ribose) synthesis promotes XRCC1 recruitment at DNA damage sites and is important for XRCC1 function.


Subject(s)
DNA Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Animals , Binding Sites , CHO Cells , Cell Line, Tumor , Cell Survival , Cricetulus , DNA Damage , Humans , Poly(ADP-ribose) Polymerases/metabolism , Protein Structure, Tertiary , X-ray Repair Cross Complementing Protein 1
11.
J Proteome Res ; 14(6): 2575-82, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25886813

ABSTRACT

Poly(ADP-ribose) polymerase 3 (PARP3) is a member of the PARP family enzymes which catalyze the ADP-ribosylation of proteins. PARP3 plays an important role in DNA damage repair and mitotic progression. In this study, we identified, using mass spectrometric techniques, two novel post-translational modification sites in PARP3, α-N-methylation and phosphorylation of serine 461 (S461). We found that the N-terminal α-amino group of PARP3 is heavily methylated in human cells, and N-terminal RCC1 methyltransferase (NRMT) is a key enzyme required for this methylation. We also observed that the phosphorylation level of S461 in PARP3 could be reduced in human cells upon treatment with flavopiridol, a cyclin-dependent kinase inhibitor. Moreover, we demonstrated that S461 phosphorylation, but not α-N-methylation of PARP3, may be involved in the cellular response toward DNA double-strand breaks. These findings provide novel insights into the post-translational regulation of PARP3.


Subject(s)
Cell Cycle Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Serine/metabolism , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Cell Line , Humans , Methylation , Phosphorylation , Poly(ADP-ribose) Polymerases/chemistry , Tandem Mass Spectrometry
12.
DNA Repair (Amst) ; 17: 30-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24680220

ABSTRACT

The repair of DNA double strand breaks is essential for cell survival and several conserved pathways have evolved to ensure their rapid and efficient repair. The non-homologous end joining pathway is initiated when Ku binds to the DNA break site. Ku is an abundant nuclear heterodimer of Ku70 and Ku80 with a toroidal structure that allows the protein to slide over the broken DNA end and bind with high affinity. Once locked into placed, Ku acts as a tool-belt to recruit multiple interacting proteins, forming one or more non-homologous end joining complexes that act in a regulated manner to ensure efficient repair of DNA ends. Here we review the structure and functions of Ku and the proteins with which it interacts during non-homologous end joining.


Subject(s)
Antigens, Nuclear/chemistry , Antigens, Nuclear/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Animals , Chromatin/metabolism , DNA Ligases/metabolism , DNA-Activated Protein Kinase/metabolism , Humans , Ku Autoantigen , Models, Molecular , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Telomere/metabolism
13.
Nucleic Acids Res ; 42(1): 307-14, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24049082

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is associated with progressive degeneration of motor neurons. Several of the genes associated with this disease encode proteins involved in RNA processing, including fused-in-sarcoma/translocated-in-sarcoma (FUS/TLS). FUS is a member of the heterogeneous nuclear ribonucleoprotein (hnRNP) family of proteins that bind thousands of pre-mRNAs and can regulate their splicing. Here, we have examined the possibility that FUS is also a component of the cellular response to DNA damage. We show that both GFP-tagged and endogenous FUS re-localize to sites of oxidative DNA damage induced by UVA laser, and that FUS recruitment is greatly reduced or ablated by an inhibitor of poly (ADP-ribose) polymerase activity. Consistent with this, we show that recombinant FUS binds directly to poly (ADP-ribose) in vitro, and that both GFP-tagged and endogenous FUS fail to accumulate at sites of UVA laser induced damage in cells lacking poly (ADP-ribose) polymerase-1. Finally, we show that GFP-FUS(R521G), harbouring a mutation that is associated with ALS, exhibits reduced ability to accumulate at sites of UVA laser-induced DNA damage. Together, these data suggest that FUS is a component of the cellular response to DNA damage, and that defects in this response may contribute to ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA Damage , Poly(ADP-ribose) Polymerases/physiology , RNA-Binding Protein FUS/metabolism , Animals , Cells, Cultured , Humans , Mice , Mutation , Oxidation-Reduction , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/biosynthesis , Poly Adenosine Diphosphate Ribose/metabolism , RNA-Binding Protein FUS/genetics
14.
DNA Repair (Amst) ; 12(8): 558-67, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23712058

ABSTRACT

A number of DNA repair disorders are known to cause neurological problems. These disorders can be broadly characterised into early developmental, mid-to-late developmental or progressive. The exact developmental processes that are affected can influence disease pathology, with symptoms ranging from early embryonic lethality to late-onset ataxia. The category these diseases belong to depends on the frequency of lesions arising in the brain, the role of the defective repair pathway, and the nature of the mutation within the patient. Using observations from patients and transgenic mice, we discuss the importance of double strand break repair during neuroprogenitor proliferation and brain development and the repair of single stranded lesions in neuronal function and maintenance.


Subject(s)
DNA Breaks, Single-Stranded , DNA Damage/genetics , Animals , Brain/growth & development , Brain/pathology , Cerebellar Ataxia/genetics , Cerebellar Ataxia/metabolism , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/metabolism , DNA Repair-Deficiency Disorders/physiopathology , Humans , Mutation , Neurogenesis/genetics , Neurons/metabolism , Neurons/pathology
15.
EMBO J ; 32(1): 112-25, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23178593

ABSTRACT

Non-homologous end joining (NHEJ) is critical for the maintenance of genetic integrity and DNA double-strand break (DSB) repair. NHEJ is regulated by a series of interactions between core components of the pathway, including Ku heterodimer, XLF/Cernunnos, and XRCC4/DNA Ligase 4 (Lig4). However, the mechanisms by which these proteins assemble into functional protein-DNA complexes are not fully understood. Here, we show that the von Willebrand (vWA) domain of Ku80 fulfills a critical role in this process by recruiting Aprataxin-and-PNK-Like Factor (APLF) into Ku-DNA complexes. APLF, in turn, functions as a scaffold protein and promotes the recruitment and/or retention of XRCC4-Lig4 and XLF, thereby assembling multi-protein Ku complexes capable of efficient DNA ligation in vitro and in cells. Disruption of the interactions between APLF and either Ku80 or XRCC4-Lig4 disrupts the assembly and activity of Ku complexes, and confers cellular hypersensitivity and reduced rates of chromosomal DSB repair in avian and human cells, respectively. Collectively, these data identify a role for the vWA domain of Ku80 and a molecular mechanism by which DNA ligase proficient complexes are assembled during NHEJ in mammalian cells, and reveal APLF to be a structural component of this critical DSB repair pathway.


Subject(s)
Antigens, Nuclear/metabolism , DNA Ligases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Antigens, Nuclear/genetics , Cell Line , Cell Survival , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair , DNA Ligase ATP , DNA Ligases/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Humans , Ku Autoantigen , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Poly-ADP-Ribose Binding Proteins , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Recombinant Fusion Proteins , Sequence Alignment , Ultraviolet Rays
16.
Mol Cell ; 41(1): 33-45, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21211721

ABSTRACT

PARP-3 is a member of the ADP-ribosyl transferase superfamily of unknown function. We show that PARP-3 is stimulated by DNA double-strand breaks (DSBs) in vitro and functions in the same pathway as the poly (ADP-ribose)-binding protein APLF to accelerate chromosomal DNA DSB repair. We implicate PARP-3 in the accumulation of APLF at DSBs and demonstrate that APLF promotes the retention of XRCC4/DNA ligase IV complex in chromatin, suggesting that PARP-3 and APLF accelerate DNA ligation during nonhomologous end-joining (NHEJ). Consistent with this, we show that class switch recombination in Aplf(-/-) B cells is biased toward microhomology-mediated end-joining, a pathway that operates in the absence of XRCC4/DNA ligase IV, and that the requirement for PARP-3 and APLF for NHEJ is circumvented by overexpression of XRCC4/DNA ligase IV. These data identify molecular roles for PARP-3 and APLF in chromosomal DNA double-strand break repair reactions.


Subject(s)
Carrier Proteins/physiology , Cell Cycle Proteins/physiology , Phosphoproteins/physiology , Poly(ADP-ribose) Polymerases/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , DNA Breaks, Double-Stranded , DNA Repair/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase , Gene Deletion , Humans , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Poly-ADP-Ribose Binding Proteins , Recombinant Fusion Proteins/physiology
17.
Mol Cell Biol ; 28(14): 4620-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18474613

ABSTRACT

APLF is a novel protein of unknown function that accumulates at sites of chromosomal DNA strand breakage via forkhead-associated (FHA) domain-mediated interactions with XRCC1 and XRCC4. APLF can also accumulate at sites of chromosomal DNA strand breaks independently of the FHA domain via an unidentified mechanism that requires a highly conserved C-terminal tandem zinc finger domain. Here, we show that the zinc finger domain binds tightly to poly(ADP-ribose), a polymeric posttranslational modification synthesized transiently at sites of chromosomal damage to accelerate DNA strand break repair reactions. Protein poly(ADP-ribosyl)ation is tightly regulated and defects in either its synthesis or degradation slow global rates of chromosomal single-strand break repair. Interestingly, APLF negatively affects poly(ADP-ribosyl)ation in vitro, and this activity is dependent on its capacity to bind the polymer. In addition, transient overexpression in human A549 cells of full-length APLF or a C-terminal fragment encoding the tandem zinc finger domain greatly suppresses the appearance of poly(ADP-ribose), in a zinc finger-dependent manner. We conclude that APLF can accumulate at sites of chromosomal damage via zinc finger-mediated binding to poly(ADP-ribose) and is a novel component of poly(ADP-ribose) signaling in mammalian cells.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , DNA Repair , Phosphoproteins/metabolism , Signal Transduction , Cell Line , DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase , Humans , Poly-ADP-Ribose Binding Proteins , Zinc Fingers
18.
Brain Res ; 1102(1): 39-43, 2006 Aug 02.
Article in English | MEDLINE | ID: mdl-16806122

ABSTRACT

Studies analysing the effects of acute treatments on animal behaviour and brain biochemistry frequently use pairwise comparisons between sham-treated and -untreated animals. In this study, we analyse expression of tPA, Grik2, Smarca2 and the transcription factor, Sp1, in mouse cerebellum following acute ethanol treatment. Expression is compared to saline-injected and -untreated control animals. We demonstrate that acute i.p. injection of saline may alter gene expression in a gene-specific manner and that ethanol may modify the effects of sham treatment on gene expression, as well as inducing specific effects independent of any handling related stress. In addition to demonstrating the complexity of gene expression in response to physical and environmental stress, this work raises questions on the interpretation and validity of studies relying on pairwise comparisons.


Subject(s)
Central Nervous System Depressants/pharmacokinetics , Cerebellum/drug effects , Ethanol/pharmacology , Gene Expression Regulation/drug effects , Handling, Psychological , Analysis of Variance , Animals , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Time Factors , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , GluK2 Kainate Receptor
19.
Mamm Genome ; 17(4): 322-31, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16596453

ABSTRACT

Analysis of the human repertoire of the FK506-binding protein (FKBP) family of peptidyl-prolyl cis/trans isomerases has identified an expansion of genes that code for human FKBPs in the secretory pathway. There are distinct differences in tissue distribution and expression levels of each variant. In this article we describe the characterization of human FKBP19 (Entrez Gene ID: FKBP11), an FK506-binding protein predominantly expressed in vertebrate secretory tissues. The FKBP19 sequence comprises a cleavable N-terminal signal sequence followed by a putative peptidyl-prolyl cis/trans isomerase domain with homology to FKBP12. This domain binds FK506 weakly in vitro. FKBP19 mRNA is abundant in human pancreas and other secretory tissues and high levels of FKBP19 protein are detected in the acinar cells of mouse pancreas.


Subject(s)
Recombinant Proteins/genetics , Tacrolimus Binding Proteins/genetics , Amino Acid Sequence , Animals , Blotting, Northern , Blotting, Western , Cattle , Cloning, Molecular , Escherichia coli/genetics , Humans , Immunoenzyme Techniques , Immunosuppressive Agents/metabolism , Mice , Mice, Inbred C3H , Molecular Sequence Data , Protein Biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tacrolimus/metabolism , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Proteins/isolation & purification , Tacrolimus Binding Proteins/metabolism , Transcription, Genetic
20.
Neurobiol Dis ; 17(2): 237-49, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15474361

ABSTRACT

The peptidyl-prolyl cis-trans isomerase (PPIase) Pin1 modulates the activity of a range of target proteins involved in the cell cycle, transcription, translation, endocytosis, and apoptosis by facilitating dephosphorylation of phosphorylated serine or threonine residue preceding a proline (p-Ser/Thr-Pro) motifs catalyzed by phosphatases specific for the trans conformations. Pin1 targets include the neuronal microtubule-associated protein tau, whose dephosphorylation restores its ability to stabilize microtubules. We, and others, have shown that tau hyperphosphorylation in the neurofibrillary tangles (NFTs) of Alzheimer disease (AD) is associated with redirection of the predominantly nuclear Pin1 to the cytoplasm and with Pin1 shortfalls throughout subcellular compartments. As nuclear Pin1 depletion causes apoptosis, shortfalls in regard to both nuclear and p-tau targets may contribute to neuronal dysfunction. We report here that similar Pin1 redistribution and shortfalls occur in frontotemporal dementias (FTDs) characterized by abnormal protein aggregates of tau and other cytoskeletal proteins. This may be a unifying, contributory factor towards neuronal death in these dementias.


Subject(s)
Brain/metabolism , Dementia/metabolism , Neurons/metabolism , Peptidylprolyl Isomerase/deficiency , Adult , Aged , Basal Ganglia , Brain/pathology , Cerebral Cortex , Dementia/pathology , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunoblotting , Immunohistochemistry , Male , Microscopy, Electron , Middle Aged , NIMA-Interacting Peptidylprolyl Isomerase , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Subcellular Fractions/metabolism , Tissue Distribution , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...