Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 78(9): 4365-4376, 2021 May.
Article in English | MEDLINE | ID: mdl-33625522

ABSTRACT

The C1ORF112 gene initially drew attention when it was found to be strongly co-expressed with several genes previously associated with cancer and implicated in DNA repair and cell cycle regulation, such as RAD51 and the BRCA genes. The molecular functions of C1ORF112 remain poorly understood, yet several studies have uncovered clues as to its potential functions. Here, we review the current knowledge on C1ORF112 biology, its evolutionary history, possible functions, and its potential relevance to cancer. C1ORF112 is conserved throughout eukaryotes, from plants to humans, and is very highly conserved in primates. Protein models suggest that C1ORF112 is an alpha-helical protein. Interestingly, homozygous knockout mice are not viable, suggesting an essential role for C1ORF112 in mammalian development. Gene expression data show that, among human tissues, C1ORF112 is highly expressed in the testes and overexpressed in various cancers when compared to healthy tissues. C1ORF112 has also been shown to have altered levels of expression in some tumours with mutant TP53. Recent screens associate C1ORF112 with DNA replication and reveal possible links to DNA damage repair pathways, including the Fanconi anaemia pathway and homologous recombination. These insights provide important avenues for future research in our efforts to understand the functions and potential disease relevance of C1ORF112.


Subject(s)
Biological Evolution , DNA Damage , DNA Repair , DNA Replication , Open Reading Frames/genetics , Animals , Humans , Male , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Interaction Maps , Testis/metabolism
2.
Front Mol Biosci ; 7: 606807, 2020.
Article in English | MEDLINE | ID: mdl-33282915

ABSTRACT

Cellular homeostasis requires adaption to environmental stress. In response to various environmental and genotoxic stresses, all cells produce dinucleoside polyphosphates (NpnNs), the best studied of which is diadenosine tetraphosphate (Ap4A). Despite intensive investigation, the precise biological roles of these molecules have remained elusive. However, recent studies have elucidated distinct and specific signaling mechanisms for these nucleotides in prokaryotes and eukaryotes. This review summarizes these key discoveries and describes the mechanisms of Ap4A and Ap4N synthesis, the mediators of the cellular responses to increased intracellular levels of these molecules and the hydrolytic mechanisms required to maintain low levels in the absence of stress. The intracellular responses to dinucleotide accumulation are evaluated in the context of the "friend" and "foe" scenarios. The "friend (or alarmone) hypothesis" suggests that ApnN act as bona fide secondary messengers mediating responses to stress. In contrast, the "foe" hypothesis proposes that ApnN and other NpnN are produced by non-canonical enzymatic synthesis as a result of physiological and environmental stress in critically damaged cells but do not actively regulate mitigating signaling pathways. In addition, we will discuss potential target proteins, and critically assess new evidence supporting roles for ApnN in the regulation of gene expression, immune responses, DNA replication and DNA repair. The recent advances in the field have generated great interest as they have for the first time revealed some of the molecular mechanisms that mediate cellular responses to ApnN. Finally, areas for future research are discussed with possible but unproven roles for intracellular ApnN to encourage further research into the signaling networks that are regulated by these nucleotides.

3.
Cell Death Dis ; 11(10): 878, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33082307

ABSTRACT

The transcriptional regulator EVI1 has an essential role in early development and haematopoiesis. However, acute myeloid leukaemia (AML) driven by aberrantly high EVI1 expression has very poor prognosis. To investigate the effects of post-translational modifications on EVI1 function, we carried out a mass spectrometry (MS) analysis of EVI1 in AML and detected dynamic phosphorylation at serine 436 (S436). Wild-type EVI1 (EVI1-WT) with S436 available for phosphorylation, but not non-phosphorylatable EVI1-S436A, conferred haematopoietic progenitor cell self-renewal and was associated with significantly higher organised transcriptional patterns. In silico modelling of EVI1-S436 phosphorylation showed reduced affinity to CtBP1, and CtBP1 showed reduced interaction with EVI1-WT compared with EVI1-S436A. The motif harbouring S436 is a target of CDK2 and CDK3 kinases, which interacted with EVI1-WT. The methyltransferase DNMT3A bound preferentially to EVI1-WT compared with EVI1-S436A, and a hypomethylated cell population associated by EVI1-WT expression in murine haematopoietic progenitors is not maintained with EVI1-S436A. These data point to EVI1-S436 phosphorylation directing functional protein interactions for haematopoietic self-renewal. Targeting EVI1-S436 phosphorylation may be of therapeutic benefit when treating EVI1-driven leukaemia.


Subject(s)
Alcohol Oxidoreductases/metabolism , Cell Self Renewal/physiology , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , MDS1 and EVI1 Complex Locus Protein/metabolism , DNA Methylation/physiology , DNA Methyltransferase 3A , DNA Modification Methylases/metabolism , Humans , Phosphorylation , Prognosis , Serine/metabolism , Transcription Factors/metabolism
4.
PLoS One ; 11(5): e0154674, 2016.
Article in English | MEDLINE | ID: mdl-27144453

ABSTRACT

Regulation of gene expression is one of several roles proposed for the stress-induced nucleotide diadenosine tetraphosphate (Ap4A). We have examined this directly by a comparative RNA-Seq analysis of KBM-7 chronic myelogenous leukemia cells and KBM-7 cells in which the NUDT2 Ap4A hydrolase gene had been disrupted (NuKO cells), causing a 175-fold increase in intracellular Ap4A. 6,288 differentially expressed genes were identified with P < 0.05. Of these, 980 were up-regulated and 705 down-regulated in NuKO cells with a fold-change ≥ 2. Ingenuity® Pathway Analysis (IPA®) was used to assign these genes to known canonical pathways and functional networks. Pathways associated with interferon responses, pattern recognition receptors and inflammation scored highly in the down-regulated set of genes while functions associated with MHC class II antigens were prominent among the up-regulated genes, which otherwise showed little organization into major functional gene sets. Tryptophan catabolism was also strongly down-regulated as were numerous genes known to be involved in tumor promotion in other systems, with roles in the epithelial-mesenchymal transition, proliferation, invasion and metastasis. Conversely, some pro-apoptotic genes were up-regulated. Major upstream factors predicted by IPA® for gene down-regulation included NFκB, STAT1/2, IRF3/4 and SP1 but no major factors controlling gene up-regulation were identified. Potential mechanisms for gene regulation mediated by Ap4A and/or NUDT2 disruption include binding of Ap4A to the HINT1 co-repressor, autocrine activation of purinoceptors by Ap4A, chromatin remodeling, effects of NUDT2 loss on transcript stability, and inhibition of ATP-dependent regulatory factors such as protein kinases by Ap4A. Existing evidence favors the last of these as the most probable mechanism. Regardless, our results suggest that the NUDT2 protein could be a novel cancer chemotherapeutic target, with its inhibition potentially exerting strong anti-tumor effects via multiple pathways involving metastasis, invasion, immunosuppression and apoptosis.


Subject(s)
Dinucleoside Phosphates/metabolism , Phosphoric Monoester Hydrolases/deficiency , Cell Line, Tumor , Down-Regulation , Gene Expression Profiling , Gene Knockout Techniques , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Phosphoric Monoester Hydrolases/genetics
5.
DNA Repair (Amst) ; 33: 90-100, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26204256

ABSTRACT

The level of intracellular diadenosine 5', 5'''-P(1),P(4)-tetraphosphate (Ap4A) increases several fold in mammalian cells treated with non-cytotoxic doses of interstrand DNA-crosslinking agents such as mitomycin C. It is also increased in cells lacking DNA repair proteins including XRCC1, PARP1, APTX and FANCG, while >50-fold increases (up to around 25 µM) are achieved in repair mutants exposed to mitomycin C. Part of this induced Ap4A is converted into novel derivatives, identified as mono- and di-ADP-ribosylated Ap4A. Gene knockout experiments suggest that DNA ligase III is primarily responsible for the synthesis of damage-induced Ap4A and that PARP1 and PARP2 can both catalyze its ADP-ribosylation. Degradative proteins such as aprataxin may also contribute to the increase. Using a cell-free replication system, Ap4A was found to cause a marked inhibition of the initiation of DNA replicons, while elongation was unaffected. Maximum inhibition of 70-80% was achieved with 20 µM Ap4A. Ap3A, Ap5A, Gp4G and ADP-ribosylated Ap4A were without effect. It is proposed that Ap4A acts as an important inducible ligand in the DNA damage response to prevent the replication of damaged DNA.


Subject(s)
DNA Damage , DNA Replication , Dinucleoside Phosphates/biosynthesis , 3T3 Cells , Adenosine Diphosphate Ribose/metabolism , Animals , CHO Cells , Cell Proliferation/drug effects , Chromatography, Ion Exchange , Cricetinae , Cricetulus , Dinucleoside Phosphates/chemistry , Epoxy Compounds/pharmacology , Gene Knockdown Techniques , HeLa Cells , Humans , Intracellular Space/metabolism , Mice , Mitomycin/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/metabolism
6.
J Biol Chem ; 289(37): 25774-82, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25070891

ABSTRACT

Fanconi anemia is a genetic disease resulting in bone marrow failure, birth defects, and cancer that is thought to encompass a defect in maintenance of genomic stability. Mutations in 16 genes (FANCA, B, C, D1, D2, E, F, G, I, J, L, M, N, O, P, and Q) have been identified in patients, with the Fanconi anemia subtype J (FA-J) resulting from homozygous mutations in the FANCJ gene. Here, we describe the direct interaction of FANCD2 with FANCJ. We demonstrate the interaction of FANCD2 and FANCJ in vivo and in vitro by immunoprecipitation in crude cell lysates and from fractions after gel filtration and with baculovirally expressed proteins. Mutation of the monoubiquitination site of FANCD2 (K561R) preserves interaction with FANCJ constitutively in a manner that impedes proper chromatin localization of FANCJ. FANCJ is necessary for FANCD2 chromatin loading and focus formation in response to mitomycin C treatment. Our results suggest not only that FANCD2 regulates FANCJ chromatin localization but also that FANCJ is necessary for efficient loading of FANCD2 onto chromatin following DNA damage caused by mitomycin C treatment.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Chromatin/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/genetics , Protein Binding , Basic-Leucine Zipper Transcription Factors/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Genomic Instability , Humans , Mutation
7.
Methods Mol Biol ; 817: 183-206, 2012.
Article in English | MEDLINE | ID: mdl-22147574

ABSTRACT

32P-postlabelling is a technique originally described by Kurt Randerath and colleagues for the sensitive detection of damage produced in DNA by reactive chemicals or genotoxins. The procedure essentially entails the enzymatic digestion of DNA to nucleoside 3'-monophosphates which are then radioactively labelled using T4 polynucleotide kinase and [γ(32)P]-adenosine triphosphate. Adducted nucleoside-3'-5'-bisphosphates are then separated from their normal counterparts by thin layer chromatography. Prior to the development of the assay, quantification of DNA adducts was confined to studies that utilised compounds synthesised to be isotopically labelled with tritium or carbon-14. As such, these studies were limited to specific and recognised genotoxins that could be administered only in the laboratory to cultures or animals. With (32)P-postlabelling it was possible not only to determine DNA adduct induction by a relatively uncharacterised suspected carcinogen, but also following exposure to complex mixtures containing a multitude of known and unknown potential genotoxins. The small amount of DNA required to perform the (32)P-postlabelling assay also meant that human biomonitoring studies using readily obtainable tissues, such as lymphocytes, were possible. Using the standard (32)P-postlabelling method, it is possible to detect a single DNA adduct in 10(7) to 10(8) normal nucleotides. The subsequent development of several enhancement methods improved this detection rate to one adduct in 10(10) nucleotides. For these reasons, the (32)-postlabelling assay represents an extremely versatile and extremely sensitive method to detect and monitor DNA damage.


Subject(s)
DNA Adducts/analysis , Mutagenicity Tests/methods , Mutagens/toxicity , Adenosine Triphosphate/chemistry , Animals , Bacteriophage T4/enzymology , Chromatography, Thin Layer/methods , DNA Adducts/genetics , DNA Adducts/metabolism , Humans , Phosphorus Radioisotopes/chemistry , Polynucleotide 5'-Hydroxyl-Kinase/chemistry
8.
Methods Mol Biol ; 817: 165-81, 2012.
Article in English | MEDLINE | ID: mdl-22147573

ABSTRACT

The single cell gel electrophoresis (SCGE) assay, more commonly known as the comet assay, due to the "comet-like" appearance of the cells, was originally developed as a technique to measure the presence of DNA single-strand breaks. The assay is performed on single cells embedded in agar and placed in an electrical field at alkaline pH, so that fragments of negatively charged single-stranded DNA move through the gel toward the positively charged anode. Undamaged DNA moves relatively slowly, forming the head of the comet, while DNA fragmented due to the presence of single-strand breaks, moves more quickly giving the appearance of the tail. The extent of DNA migration is a measure of the DNA damage present. Since it was first developed, the comet assay has been adapted for measuring other types of DNA damage. The neutral comet assay has been employed for DNA double-strand breaks, while techniques using DNA repair enzymes to cleave specific adducts, UvrABC for ultraviolet radiation induced adducts, for example, have also been described. Here, we describe a modified version of the comet assay for the measurement of interstrand crosslinks (ICLs). Interstrand crosslinking agents include the chemotherapeutic agents mitomycin C and cis-platin, psoralen plus UVA light (PUVA) used to treat hyperproliferative skin disorders and diepoxybutane, a metabolite of 1,3-butadiene used in industrial processes and an environmental pollutant. ICLs are a potent and cytotoxic form of DNA damage as they prevent DNA strand separation, thereby preventing DNA replication. Their removal requires several different DNA repair processes including translesion synthesis and homologous recombination. As ICLs prevent separation of the DNA strands, their presence results in less DNA migration in the comet assay. To successfully measure ICLs, it is necessary to incorporate a step that induces single-strand breaks (using a defined dose of ionizing radiation) that allows the crosslinked DNA to migrate.


Subject(s)
Comet Assay/methods , DNA Damage/drug effects , DNA/genetics , Mutagens/toxicity , Animals , Cell Line , Cross-Linking Reagents/toxicity , DNA/analysis , DNA Damage/radiation effects , Epoxy Compounds/toxicity , Ficusin/toxicity , Gamma Rays , Humans , Mitomycin/toxicity , Ultraviolet Rays
9.
Am J Pathol ; 179(2): 1061-72, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21801876

ABSTRACT

Two subgroups of invasive breast carcinomas have been identified with a poor prognosis in different patient cohorts: the basal-like category and the subgroup containing proteins capable of inducing metastasis in experimental rodents, the metastasis-inducing proteins (MIPs). Here we identify by immunohistochemical staining for cytokeratin CK5/6 or CK14 the basal-like subgroup in a set of 297 primary invasive breast carcinomas in which the staining profile for the MIPs S100A4, osteopontin, anterior gradient-2, and S100P has already been established. Monoclonal antibodies to CK5/6 or CK14 specifically stain 31% to 34% of the primary carcinomas. These positively stained tumors are highly significantly associated with premature death of the patient (Wilcoxon statistics, P < 0.0001), the increased relative risk being approximately 5.6-fold. Positive staining for either cytokeratin is very significantly associated with that for each of the four MIPs separately and with loss of staining for the Fanconi anemia protein FANCD2 (corrected Fisher's exact test, P < 0.0007). There is no significant correlation with the remaining tumor variables tested, including staining for the estrogen receptor α, progesterone receptor, and c-erbB-2. These results show that the basal cytokeratin-like carcinomas contain many of the MIPs and that these may arise by their selection for tumors with an inherent deficiency in the FANC/BRCA pathway of DNA repair.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Keratins/metabolism , Neoplasms, Basal Cell/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry/methods , Lymph Nodes/pathology , Middle Aged , Models, Statistical , Neoplasm Metastasis , Retrospective Studies , Time Factors
10.
Hum Mol Genet ; 20(22): 4395-410, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21865299

ABSTRACT

Fanconi anemia (FA) is a rare genetic disorder characterized by bone marrow failure and an increased risk for leukemia and cancer. Fifteen proteins thought to function in the repair of DNA interstrand crosslinks (ICLs) comprise what is known as the FA-BRCA pathway. Activation of this pathway leads to the monoubiquitylation and chromatin localization of FANCD2 and FANCI. It has previously been shown that FANCJ interacts with the mismatch repair (MMR) complex MutLα. Here we show that FANCD2 interacts with the MMR proteins MSH2 and MLH1. FANCD2 monoubiquitylation, foci formation and chromatin loading are greatly diminished in MSH2-deficient cells. Human or mouse cells lacking MSH2 or MLH1 display increased sensitivity and radial formation in response to treatment with DNA crosslinking agents. Studies in human cell lines and Drosophila mutants suggest an epistatic relationship between FANCD2, MSH2 and MLH1 with regard to ICL repair. Surprisingly, the interaction between MSH2 and MLH1 is compromised in multiple FA cell lines, and FA cell lines exhibit deficient MMR. These results suggest a significant role for MMR proteins in the activation of the FA pathway and repair of ICLs. In addition, we provide the first evidence for a defect in MMR in FA cell lines.


Subject(s)
DNA Mismatch Repair/physiology , Fanconi Anemia/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Drosophila , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , HCT116 Cells , HeLa Cells , Humans , Mice , MutL Protein Homolog 1 , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Signal Transduction/genetics , Signal Transduction/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
Mutat Res ; 689(1-2): 12-20, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20450923

ABSTRACT

The Fanconi anaemia (FA) FANCG protein is an integral component of the FA nuclear core complex that is required for monoubiquitylation of FANCD2. FANCG is also part of another protein complex termed D1-D2-G-X3 that contains FANCD2 and the homologous recombination repair proteins BRCA2 (FANCD1) and XRCC3. Formation of the D1-D2-G-X3 complex is mediated by serine-7 phosphorylation of FANCG and occurs independently of the FA core complex and FANCD2 monoubiquitylation. FANCG contains seven tetratricopeptide repeat (TPR) motifs that mediate protein-protein interactions and here we show that mutation of several of the TPR motifs at a conserved consensus residue ablates the in vivo binding activity of FANCG. Expression of mutated TPR1, TPR2, TPR5 and TPR6 in Chinese hamster fancg mutant NM3 fails to functionally complement its hypersensitivities to mitomycin C (MMC) and phleomycin and fails to restore FANCD2 monoubiquitylation. Using co-immunoprecipitation analysis, we demonstrate that these TPR-mutated FANCG proteins fail to interact with BRCA2, XRCC3, FANCA or FANCF. The interactions of other proteins in the D1-D2-G-X3 complex are also absent, including the interaction of BRCA2 with both the monoubiquitylated (FANCD2-L) and non-ubiquitylated (FANCD2-S) isoforms of FANCD2. Interestingly, a mutation of TPR7 (R563E), that complements the MMC and phleomycin hypersensitivity of human FA-G EUFA316 cells, fails to complement NM3, despite the mutated FANCG protein co-precipitating with FANCA, BRCA2 and XRCC3. Whilst interaction of TPR7-mutated FANCG with FANCF does appear to be reduced in NM3, FANCD2 is monoubiquitylated suggesting that sub-optimal interactions of FANCG in the core complex and the D1-D2-G-X3 complex are responsible for the observed MMC- and phleomycin-hypersensitivity, rather than a defect in FANCD2 monoubiquitylation. Our data demonstrate that FANCG functions as a mediator of protein-protein interactions and is vital for the assembly of multi-protein complexes including the FA core complex and the D1-D2-G-X3 complex.


Subject(s)
Amino Acid Motifs , BRCA2 Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group G Protein/chemistry , Nucleic Acid Synthesis Inhibitors/pharmacology , Phleomycins/pharmacology , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , DNA-Binding Proteins/metabolism , Humans , Mice , Mutation , Recombination, Genetic , Transfection , Ubiquitination
12.
Am J Pathol ; 176(6): 2935-47, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20363922

ABSTRACT

FANCD2, a pivotal protein in the Fanconi anemia and BRCA pathway/network, is monoubiquitylated in the nucleus in response to DNA damage. This study examines the subcellular location and relationship with prognostic factors and patient survival of FANCD2 in breast cancer. Antibodies to FANCD2 were used to immunocytochemically stain 16 benign and 20 malignant breast specimens as well as 314 primary breast carcinomas to assess its association with subcellular compartment and prognostic factors using Fisher's Exact test or with patient survival over 20 years using Wilcoxon-Gehan statistics. Immunoreactive FANCD2 was found in the nucleus and cytoplasm of all 16 benign tissues, but nuclear staining was lost from a significant 19/20 malignant carcinomas (P < 0.0001). Antibodies to FANCD2 stained the cytoplasm of 196 primary carcinomas, leaving 118 as negatively stained. Negative cytoplasmic staining was significantly associated with positive staining for the metastasis-inducing proteins S100A4, S100P, osteopontin, and AGR2 (P < or = 0.002). Survival of patients with FANCD2-negative carcinomas was significantly worse (P < 0.0001) than those with positively stained carcinomas, and only 4% were alive at the census date. Multivariate regression analysis identified negative staining for cytoplasmic FANCD2 as the most significant indicator of patient death (P = 0.001). Thus FANCD2's cytoplasmic loss in the primary carcinomas may allow the selection of cells overexpressing proteins that can induce metastases before surgery.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fanconi Anemia Complementation Group D2 Protein/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/diagnosis , Breast Neoplasms/mortality , Cells, Cultured , Female , Humans , Middle Aged , Neoplasm Metastasis , Prognosis , Survival Rate
13.
Mol Cell ; 37(6): 749-51, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20347418

ABSTRACT

In this issue of Molecular Cell, Yan et al. (2010) and Singh et al. (2010) identify an evolutionarily conserved FANCM-associated histone-fold MHF heterodimer (MHF1-MHF2) that promotes the remodeling of artificial replication forks and confers cellular resistance to DNA crosslinks and camptothecin.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA Replication , Fanconi Anemia/metabolism , Genomic Instability , DNA Helicases/genetics , Evolution, Molecular , Fanconi Anemia/genetics , Humans , Protein Multimerization , Signal Transduction
14.
Cancer Res ; 69(22): 8775-83, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19861535

ABSTRACT

Fanconi anemia is a cancer-prone inherited bone marrow failure and cancer susceptibility syndrome with at least 13 complementation groups (FANCA, FANCB, FANCC, FANCD1, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCJ, FANCL, FANCM, and FANCN). Our laboratory has previously described several regulatory phosphorylation events for core complex member proteins FANCG and FANCA by phosphorylation. In this study, we report a novel phosphorylation site serine 331 (S331) of FANCD2, the pivotal downstream player of the Fanconi anemia pathway. Phosphorylation of S331 is important for its DNA damage-inducible monoubiquitylation, resistance to DNA cross-linkers, and in vivo interaction with FANCD1/BRCA2. A phosphomimetic mutation at S331 restores all of these phenotypes to wild-type. In vitro and in vivo experiments show that phosphorylation of S331 is mediated by CHK1, the S-phase checkpoint kinase implicated in the Fanconi anemia DNA repair pathway.


Subject(s)
BRCA2 Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Cell Line , Checkpoint Kinase 1 , DNA Damage/physiology , Fanconi Anemia Complementation Group Proteins/metabolism , Humans , Immunoblotting , Immunoprecipitation , Mass Spectrometry , Phosphorylation , Protein Kinases/metabolism , Sequence Homology, Amino Acid , Serine
15.
Mutagenesis ; 24(2): 173-81, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19147795

ABSTRACT

Psoralen plus ultraviolet A (PUVA), commonly used for the treatment of hyperproliferative skin disorders, has been found to be associated with an increased risk of squamous cell cancer. Interstrand cross-link (ICL) formation by PUVA treatment is considered the major factor contributing to the carcinogenesis. However, it remains unclear how PUVA causes, or promotes cancers, in humans. As an initial step in understanding the mechanisms of mutagenesis and carcinogenesis of PUVA photochemotherapy, we have optimized and subsequently utilized a modified alkaline comet assay involving a post-lysis gamma-irradiation at 9 Gy to sensitively measure the formation and repair of PUVA-induced ICLs in the immortalized human keratinocyte cell line HaCaT. A clear dose-dependent response of HaCaT cells to PUVA exposure was observed with a combination of a fixed UVA dose at 0.05 J/cm(2) and a dose of 8-methoxypsoralen ranging from 10 to 100 microM. Results also indicated that the ICL repair was concentration dependent. We have also demonstrated that PUVA-induced monoadduct formation, at an estimated ratio of 3:1 to ICLs in the present experimental conditions, does not interfere with the detection of the ICLs in the modified alkaline comet assay. Furthermore, comparison of the amount of ICL formation between the single-dose UVA treatment and a split-dose protocol was performed. The split-dose protocol was believed to generate more ICLs than the single-dose treatment, thus more effective in PUVA photochemotherapy. Our results demonstrate that comparable amounts of ICLs were formed in HaCaT cells for each dose of UVA, using either the split-dose or single-dose protocols.


Subject(s)
Comet Assay/methods , Cross-Linking Reagents/metabolism , DNA/metabolism , PUVA Therapy , Cell Line , DNA Adducts/chemistry , DNA Adducts/drug effects , DNA Adducts/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Dose-Response Relationship, Radiation , Furocoumarins/chemistry , Furocoumarins/pharmacology , Gamma Rays , Humans , Keratinocytes/drug effects , Keratinocytes/radiation effects , Methoxsalen/pharmacology
16.
Blood ; 113(10): 2181-90, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19109555

ABSTRACT

Previous work has shown several proteins defective in Fanconi anemia (FA) are phosphorylated in a functionally critical manner. FANCA is phosphorylated after DNA damage and localized to chromatin, but the site and significance of this phosphorylation are unknown. Mass spectrometry of FANCA revealed one phosphopeptide, phosphorylated on serine 1449. Serine 1449 phosphorylation was induced after DNA damage but not during S phase, in contrast to other posttranslational modifications of FA proteins. Furthermore, the S1449A mutant failed to completely correct a variety of FA-associated phenotypes. The DNA damage response is coordinated by phosphorylation events initiated by apical kinases ATM (ataxia telangectasia mutated) and ATR (ATM and Rad3-related), and ATR is essential for proper FA pathway function. Serine 1449 is in a consensus ATM/ATR site, phosphorylation in vivo is dependent on ATR, and ATR phosphorylated FANCA on serine 1449 in vitro. Phosphorylation of FANCA on serine 1449 is a DNA damage-specific event that is downstream of ATR and is functionally important in the FA pathway.


Subject(s)
Cell Cycle Proteins/metabolism , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Protein Serine-Threonine Kinases/metabolism , Alkylating Agents/toxicity , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , DNA Damage/drug effects , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Immunoprecipitation , Mass Spectrometry , Mitomycin/toxicity , Phosphorylation , Serine/metabolism
17.
DNA Repair (Amst) ; 5(5): 629-40, 2006 May 10.
Article in English | MEDLINE | ID: mdl-16621732

ABSTRACT

Fanconi anaemia is an inherited chromosomal instability disorder characterised by cellular sensitivity to DNA interstrand crosslinkers, bone-marrow failure and a high risk of cancer. Eleven FA genes have been identified, one of which, FANCD1, is the breast cancer susceptibility gene BRCA2. At least eight FA proteins form a nuclear core complex required for monoubiquitination of FANCD2. The BRCA2/FANCD1 protein is connected to the FA pathway by interactions with the FANCG and FANCD2 proteins, both of which co-localise with the RAD51 recombinase, which is regulated by BRCA2. These connections raise the question of whether any of the FANC proteins of the core complex might also participate in other complexes involved in homologous recombination repair. We therefore tested known FA proteins for direct interaction with RAD51 and its paralogs XRCC2 and XRCC3. FANCG was found to interact with XRCC3, and this interaction was disrupted by the FA-G patient derived mutation L71P. FANCG was co-immunoprecipitated with both XRCC3 and BRCA2 from extracts of human and hamster cells. The FANCG-XRCC3 and FANCG-BRCA2 interactions did not require the presence of other FA proteins from the core complex, suggesting that FANCG also participates in a DNA repair complex that is downstream and independent of FANCD2 monoubiquitination. Additionally, XRCC3 and BRCA2 proteins co-precipitate in both human and hamster cells and this interaction requires FANCG. The FANCG protein contains multiple tetratricopeptide repeat motifs (TPRs), which function as scaffolds to mediate protein-protein interactions. Mutation of one or more of these motifs disrupted all of the known interactions of FANCG. We propose that FANCG, in addition to stabilising the FA core complex, may have a role in building multiprotein complexes that facilitate homologous recombination repair.


Subject(s)
BRCA2 Protein/metabolism , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group G Protein/metabolism , Amino Acid Motifs , Animals , BRCA2 Protein/genetics , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , DNA Repair , DNA-Binding Proteins/genetics , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group G Protein/chemistry , Fanconi Anemia Complementation Group G Protein/genetics , HeLa Cells , Humans , In Vitro Techniques , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombination, Genetic , Two-Hybrid System Techniques
18.
Environ Mol Mutagen ; 45(2-3): 128-42, 2005.
Article in English | MEDLINE | ID: mdl-15668941

ABSTRACT

The genetically complex disease Fanconi anemia (FA) comprises cancer predisposition, developmental defects, and bone marrow failure due to elevated apoptosis. The FA cellular phenotype includes universal sensitivity to DNA crosslinking damage, symptoms of oxidative stress, and reduced mutability at the X-linked HPRT gene. In this review article, we present a new heuristic molecular model that accommodates these varied features of FA cells. In our view, the FANCA, -C, and -G proteins, which are both cytoplasmic and nuclear, have an integrated dual role in which they sense and convey information about cytoplasmic oxidative stress to the nucleus, where they participate in the further assembly and functionality of the nuclear core complex (NCCFA= FANCA/B/C/E/F/G/L). In turn, NCCFA facilitates DNA replication at sites of base damage and strand breaks by performing the critical monoubiquitination of FANCD2, an event that somehow helps stabilize blocked and broken replication forks. This stabilization facilitates two kinds of processes: translesion synthesis at sites of blocking lesions (e.g., oxidative base damage), which produces point mutations by error-prone polymerases, and homologous recombination-mediated restart of broken forks, which arise spontaneously and when crosslinks are unhooked by the ERCC1-XPF endonuclease. In the absence of the critical FANCD2 monoubiquitination step, broken replication forks further lose chromatid continuity by collapsing into a configuration that is more difficult to restart through recombination and prone to aberrant repair through nonhomologous end joining. Thus, the FA regulatory pathway promotes chromosome integrity by monitoring oxidative stress and coping efficiently with the accompanying oxidative DNA damage during DNA replication.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , DNA Repair/physiology , DNA Replication/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Models, Molecular , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Signal Transduction/physiology , Cell Cycle Proteins/genetics , Cell Nucleus/metabolism , Cell Nucleus/physiology , Chromatin/metabolism , Cytoplasm/metabolism , DNA Damage , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group Proteins , Mutation/genetics , Nuclear Proteins/genetics , Oxidative Stress/genetics , Oxidative Stress/physiology , Ubiquitins/metabolism
19.
DNA Repair (Amst) ; 4(1): 11-22, 2005 Jan 02.
Article in English | MEDLINE | ID: mdl-15533833

ABSTRACT

The Fanconi anemia (FA) proteins overlap with those of homologous recombination through FANCD1/BRCA2, but the biochemical functions of other FA proteins are largely unknown. By constructing and characterizing a null fancg mutant (KO40) of hamster CHO cells, we show that FancG protects cells against a broad spectrum of genotoxic agents. KO40 is consistently hypersensitive to both alkylating agents that produce monoadducts and those that produce interstrand crosslinks. KO40 cells were no more sensitive to mitomycin C (3x) and diepoxybutane (2x) than to 6-thioguanine (5x), ethylnitrosourea (3x), or methyl methanesulfonate (MMS) (3x). These results contrast with the pattern of selective sensitivity to DNA crosslinking agents seen historically with cell lines from FA patients. The hypersensitivity of KO40 to MMS was not associated with a higher level of initial DNA single-strand breaks; nor was there a defect in removing MNU-induced methyl groups from DNA. Both control and MMS-treated synchronized G1-phase KO40 cells progressed through S phase at a normal rate but showed a lengthening of G2 phase compared with wild type. MMS-treated and untreated early S-phase KO40 cells had increased levels of Rad51 foci compared with wild type. Asynchronous KO40 treated with ionizing radiation (IR) exhibited a normal Rad51 focus response, consistent with KO40 having only slight sensitivity to killing by IR. The plating efficiency and doubling time of KO40 cells were nearly normal, and they showed no increase in spontaneous chromosomal aberrations or sister chromatid exchanges. Collectively, our results do not support a role for FancG during DNA replication that deals specifically with processing DNA crosslinks. Nor do they suggest that the main function of the FA protein "pathway" is to promote efficient homologous recombination. We propose that the primary function of FA proteins is to maintain chromosomal continuity by stabilizing replication forks that encounter nicks, gaps, or replication-blocking lesions.


Subject(s)
DNA Repair , DNA Replication/drug effects , DNA-Binding Proteins/genetics , Fanconi Anemia/metabolism , Mutation/genetics , Animals , CHO Cells , Cell Cycle/drug effects , Cricetinae , Cricetulus , Cross-Linking Reagents/toxicity , Cytoprotection/genetics , DNA Replication/physiology , DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group G Protein , Fluorescent Antibody Technique , Gene Targeting , Methyl Methanesulfonate/toxicity , Mutagens/toxicity , Plasmids/genetics , Rad51 Recombinase , Radiation, Ionizing , Transfection
20.
Radiat Res ; 162(6): 667-76, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15548117

ABSTRACT

The clonogenic survival of cells of the radiation-sensitive hamster cell lines irs1, irs2, irs3 and xrs5, representing different DNA repair pathways, was compared to that of their parent lines after alpha-particle irradiation. Measurements of nuclear area were made to calculate the probability of surviving a single alpha-particle traversal, the average number of lethal lesions per track and per unit dose, along with the "intrinsic radiosensitivity" of these cells, allowing for the potential of multiple lethal lesions per traversal. For all cell lines studied, alpha particles were found to be more biologically effective per unit absorbed dose than X rays at inducing cell inactivation. The repair-deficient cells showed an enhanced sensitivity to alpha particles compared to their parent line, but the degree of enhancement was less than for X rays. The reduction in additional sensitivity for alpha-particle irradiation was shown not to be due predominantly to differences in cell geometry limiting the probability of a cell nucleus being traversed. The results suggest that both the nonhomologous end-joining pathway and to a lesser extent the homologous recombination repair pathway play a role in successful repair of alpha-particle-induced damage, although a large proportion of damage is not repaired by either pathway.


Subject(s)
Alpha Particles , DNA Repair , Animals , CHO Cells , Cell Survival/radiation effects , Cricetinae , DNA Damage , Linear Energy Transfer , Radiation Tolerance , Relative Biological Effectiveness , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...