Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Biochem ; 166(4): 343-351, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31119278

ABSTRACT

Matrin3 is a highly conserved inner nuclear matrix protein involved in multiple stages of RNA metabolism. Although Matrin3 may also play a role in DNA repair, its precise roles have remained unclear. In this study, we showed that the depletion of Matrin3 led to decreased homologous recombination (HR) efficiency and increased radiation sensitivity of cells. Matrin3-depleted cells showed impaired DNA damage-dependent focus formation of RAD51, a key protein in HR. These findings suggest that Matrin3 promotes HR by regulating RAD51.

2.
Elife ; 72018 05 08.
Article in English | MEDLINE | ID: mdl-29759113

ABSTRACT

Chromosomal translocations are hallmarks of various types of cancers and leukemias. However, the molecular mechanisms of chromosome translocations remain largely unknown. The ataxia-telangiectasia mutated (ATM) protein, a DNA damage signaling regulator, facilitates DNA repair to prevent chromosome abnormalities. Previously, we showed that ATM deficiency led to the 11q23 chromosome translocation, the most frequent chromosome abnormalities in secondary leukemia. Here, we show that ARP8, a subunit of the INO80 chromatin remodeling complex, is phosphorylated after etoposide treatment. The etoposide-induced phosphorylation of ARP8 is regulated by ATM and ATR, and attenuates its interaction with INO80. The ATM-regulated phosphorylation of ARP8 reduces the excessive loading of INO80 and RAD51 onto the breakpoint cluster region. These findings suggest that the phosphorylation of ARP8, regulated by ATM, plays an important role in maintaining the fidelity of DNA repair to prevent the etoposide-induced 11q23 abnormalities.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Helicases/metabolism , Microfilament Proteins/metabolism , Protein Processing, Post-Translational , Translocation, Genetic , ATPases Associated with Diverse Cellular Activities , Cell Line , DNA Repair , DNA-Binding Proteins , Etoposide/toxicity , Humans , Phosphorylation , Rad51 Recombinase/metabolism
3.
Hypertens Res ; 41(6): 426-434, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29626209

ABSTRACT

Deficiency of X-ray repair cross-complementing protein 3 (XRCC3), a DNA-damage repair molecule, and the 241Met variant of XRCC3 have been reported to increase endoreduplication, which induces polyploidy. The aims of this study were to determine the impact of the XRCC3 polymorphism on the incidence of hypertension-induced left ventricular hypertrophy (LVH) and to investigate the mechanisms underlying any potential relationship. Patients undergoing chronic hemodialysis (n = 77) were genotyped to assess for the XRCC3 Thr241Met polymorphism. The XRCC3 241Thr/Met genotype was more frequent in the LVH (+) group than in the LVH (-) group (42.3 vs. 13.7%, χ2 = 7.85, p = 0.0051). To investigate possible mechanisms underlying these observations, human XRCC3 cDNA of 241Thr or that of 241Met was introduced into cultured CHO cells. The surface area of CHO cells expressing XRCC3 241Met was larger than that expressing 241Thr. Spontaneous DNA double-strand breaks accumulated to a greater degree in NIH3T3 cells expressing 241Met (3T3-241Met) than in those expressing 241Thr (3T3-241Thr). DNA damage caused by radiation induced cell senescence more frequently in 3T3-241Met. The levels of basal and TNF-α-stimulated MCP-1 mRNA and protein secretion were higher in 3T3-241Met. Finally, FACS analysis revealed that the cell percentage in G2/M phase including polyploidy was significantly higher in 3T3-241Met than in 3T3-241Thr. Furthermore, the basal level of MCP-1 mRNA positively correlated with the cell percentage in G2/M phase and polyploidy. These data suggest that the XRCC3 241Met increases the risk of LVH via accumulation of DNA damage, thereby altering cell cycle progression and inducing cell senescence and a proinflammatory phenotype.


Subject(s)
DNA-Binding Proteins/genetics , Hypertension/complications , Hypertrophy, Left Ventricular/genetics , Aged , Aged, 80 and over , Animals , CHO Cells , Cricetulus , Female , Humans , Male , Mice , Middle Aged , NIH 3T3 Cells
4.
Cancer Sci ; 107(4): 444-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26825989

ABSTRACT

The epidermal growth factor receptor (EGFR) tyrosine kinase signaling pathways regulate cellular activities. The EGFR tyrosine kinase inhibitors (EGFR-TKIs) repress the EGFR pathway constitutively activated by somatic EGFR gene mutations and have drastically improved the prognosis of non-small-cell lung cancer (NSCLC) patients. However, some problems, including resistance, remain to be solved. Recently, combination therapy with EGFR-TKIs and cytotoxic agents has been shown to improve the prognosis of NSCLC patients. To enhance the anticancer effects of EGFR-TKIs, we examined the cross-talk of the EGFR pathways with ataxia telangiectasia-mutated (ATM) signaling pathways. ATM is a key protein kinase in the DNA damage response and is known to phosphorylate Akt, an EGFR downstream factor. We found that the combination of an ATM inhibitor, KU55933, and an EGFR-TKI, gefitinib, resulted in synergistic cell growth inhibition and induction of apoptosis in NSCLC cell lines carrying the sensitive EGFR mutation. We also found that KU55933 enhanced the gefitinib-dependent repression of the phosphorylation of EGFR and/or its downstream factors. ATM inhibition may facilitate the gefitinib-dependent repression of the phosphorylation of EGFR and/or its downstream factors, to exert anticancer effects against NSCLC cells with the sensitive EGFR mutation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/biosynthesis , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , Gefitinib , Humans , Morpholines/administration & dosage , Mutation , Protein Kinase Inhibitors/administration & dosage , Pyrones/administration & dosage , Quinazolines/administration & dosage , Signal Transduction/drug effects
5.
Biochemistry ; 55(4): 637-46, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26757249

ABSTRACT

Linker histones bind to nucleosomes and compact polynucleosomes into a higher-order chromatin configuration. Somatic and germ cell-specific linker histone subtypes have been identified and may have distinct functions. In this study, we reconstituted polynucleosomes containing human histones H1.2 and H1T, as representative somatic and germ cell-specific linker histones, respectively, and found that H1T forms less compacted chromatin, as compared to H1.2. An in vitro homologous pairing assay revealed that H1T weakly inhibited RAD51/RAD54-mediated homologous pairing in chromatin, although the somatic H1 subtypes, H1.0, H1.1, H1.2, H1.3, H1.4, and H1.5, substantially suppressed it. An in vivo recombination assay revealed that H1T overproduction minimally affected the recombination frequency, but significant suppression was observed when H1.2 was overproduced in human cells. These results suggested that the testis-specific linker histone, H1T, possesses a specific function to produce the chromatin architecture required for proper chromosome regulation, such as homologous recombination.


Subject(s)
DNA Helicases/chemistry , Histones/chemistry , Nuclear Proteins/chemistry , Nucleosomes/chemistry , Rad51 Recombinase/chemistry , Recombination, Genetic , Cell Line , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins , Histones/genetics , Histones/immunology , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
6.
Int J Radiat Oncol Biol Phys ; 89(4): 736-44, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24969791

ABSTRACT

PURPOSE: The reorganization of damaged chromatin plays an important role in the regulation of the DNA damage response. A recent study revealed the presence of 2 vertebrate H2A.Z isoforms, H2A.Z-1 and H2A.Z-2. However, the roles of the vertebrate H2A.Z isoforms are still unclear. Thus, in this study we examined the roles of the vertebrate H2A.Z isoforms in chromatin reorganization after the induction of DNA double-strand breaks (DSBs). METHODS AND MATERIALS: To examine the dynamics of H2A.Z isoforms at damaged sites, we constructed GM0637 cells stably expressing each of the green fluorescent protein (GFP)-labeled H2A.Z isoforms, and performed fluorescence recovery after photobleaching (FRAP) analysis and inverted FRAP analysis in combination with microirradiation. Immunofluorescence staining using an anti-RAD51 antibody was performed to study the kinetics of RAD51 foci formation after 2-Gy irradiation of wild-type (WT), H2A.Z-1- and H2A.Z-2-deficient DT40 cells. Colony-forming assays were also performed to compare the survival rates of WT, H2A.Z-1-, and H2A.Z-2-deficient DT40 cells with control, and H2A.Z-1- and H2A.Z-2-depleted U2OS cells after irradiation. RESULTS: FRAP analysis revealed that H2A.Z-2 was incorporated into damaged chromatin just after the induction of DSBs, whereas H2A.Z-1 remained essentially unchanged. Inverted FRAP analysis showed that H2A.Z-2 was released from damaged chromatin. These findings indicated that H2A.Z-2 was exchanged at DSB sites immediately after the induction of DSBs. RAD51 focus formation after ionizing irradiation was disturbed in H2A.Z-2-deficient DT40 cells but not in H2A.Z-1-deficient cells. The survival rate of H2A.Z-2-deficient cells after irradiation was lower than those of WT and H2A.Z-1- DT40 cells. Similar to DT40 cells, H2A.Z-2-depleted U2OS cells were also radiation-sensitive compared to control and H2A.Z-1-depleted cells. CONCLUSIONS: We found that vertebrate H2A.Z-2 is involved in the regulation of the DNA damage response at a very early stage, via the damaged chromatin reorganization required for RAD51 focus formation.


Subject(s)
Chromatin/metabolism , DNA Breaks, Double-Stranded , DNA Repair/physiology , Histones/metabolism , Rad51 Recombinase/metabolism , Cell Survival/physiology , Cells, Cultured , Chromatin/chemistry , Chromatin/genetics , Colony-Forming Units Assay/methods , Fluorescent Antibody Technique/methods , Histones/genetics , Humans , Protein Isoforms/metabolism
7.
Sci Rep ; 4: 4863, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24798879

ABSTRACT

Homologous recombination plays essential roles in mitotic DNA double strand break (DSB) repair and meiotic genetic recombination. In eukaryotes, RAD51 promotes the central homologous-pairing step during homologous recombination, but is not sufficient to overcome the reaction barrier imposed by nucleosomes. RAD54, a member of the ATP-dependent nucleosome remodeling factor family, is required to promote the RAD51-mediated homologous pairing in nucleosomal DNA. In higher eukaryotes, most nucleosomes form higher-ordered chromatin containing the linker histone H1. However, the mechanism by which RAD51/RAD54-mediated homologous pairing occurs in higher-ordered chromatin has not been elucidated. In this study, we found that a histone chaperone, Nap1, accumulates on DSB sites in human cells, and DSB repair is substantially decreased in Nap1-knockdown cells. We determined that Nap1 binds to RAD54, enhances the RAD54-mediated nucleosome remodeling by evicting histone H1, and eventually stimulates the RAD51-mediated homologous pairing in higher-ordered chromatin containing histone H1.


Subject(s)
Chromatin/metabolism , DNA Helicases/metabolism , Histones/metabolism , Homologous Recombination/genetics , Nuclear Proteins/metabolism , Proteins/metabolism , Rad51 Recombinase/metabolism , Adenosine Triphosphatases/metabolism , Cell Line , Chromatin/genetics , DNA Helicases/genetics , DNA Repair/genetics , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Histones/genetics , Humans , Nuclear Proteins/genetics , Nucleosomes/genetics , Nucleosomes/metabolism , Rad51 Recombinase/genetics , tRNA Methyltransferases
8.
J Cell Sci ; 126(Pt 22): 5284-92, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24046452

ABSTRACT

Genetic information encoded in chromosomal DNA is challenged by intrinsic and exogenous sources of DNA damage. DNA double-strand breaks (DSBs) are extremely dangerous DNA lesions. RAD51 plays a central role in homologous DSB repair, by facilitating the recombination of damaged DNA with intact DNA in eukaryotes. RAD51 accumulates at sites containing DNA damage to form nuclear foci. However, the mechanism of RAD51 accumulation at sites of DNA damage is still unclear. Post-translational modifications of proteins, such as phosphorylation, acetylation and ubiquitylation play a role in the regulation of protein localization and dynamics. Recently, the covalent binding of small ubiquitin-like modifier (SUMO) proteins to target proteins, termed SUMOylation, at sites containing DNA damage has been shown to play a role in the regulation of the DNA-damage response. Here, we show that the SUMOylation E2 ligase UBC9, and E3 ligases PIAS1 and PIAS4, are required for RAD51 accretion at sites containing DNA damage in human cells. Moreover, we identified a SUMO-interacting motif (SIM) in RAD51, which is necessary for accumulation of RAD51 at sites of DNA damage. These findings suggest that the SUMO-SIM system plays an important role in DNA repair, through the regulation of RAD51 dynamics.


Subject(s)
DNA Damage/genetics , DNA Repair/genetics , Rad51 Recombinase/genetics , Sumoylation/genetics , Cell Line , DNA Breaks, Double-Stranded , Humans , Poly-ADP-Ribose Binding Proteins , Protein Inhibitors of Activated STAT/metabolism , Protein Processing, Post-Translational/genetics , Rad51 Recombinase/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
9.
Radiat Res ; 177(5): 533-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22509803

ABSTRACT

The analysis of dicentric chromosomes in human peripheral blood lymphocytes (PBLs) by Giemsa staining is the most established method for biological dosimetry. However, this method requires a well-trained person because of the difficulty in detecting aberrations rapidly and accurately. Here, we applied a fluorescence in situ hybridization (FISH) technique, using telomere and centromere peptide nucleic acid (PNA) probes, to solve the problem of biological dosimetry in radiation emergency medicine. A comparison by a well-trained observer found that FISH analysis of PBLs for the dose estimation was more accurate than the conventional Giemsa analysis, especially in samples irradiated at high doses. These results show that FISH analysis with centromeric/telomeric PNA probes could become the standard method for biological dosimetry in radiation emergency medicine.


Subject(s)
Chromosome Aberrations/radiation effects , Chromosomes, Human/radiation effects , In Situ Hybridization, Fluorescence/methods , Molecular Probes , Peptide Nucleic Acids , Radiometry/methods , Adult , Azure Stains , Centromere/ultrastructure , Chromosome Breakage/radiation effects , Chromosomes, Human/ultrastructure , Dose-Response Relationship, Radiation , Emergency Medicine/methods , Female , Gamma Rays/adverse effects , Humans , In Vitro Techniques , Lymphocytes/radiation effects , Lymphocytes/ultrastructure , Male , Metaphase , Middle Aged , Peptide Nucleic Acids/genetics , Ring Chromosomes , Staining and Labeling , Telomere/ultrastructure
10.
EMBO Rep ; 13(1): 44-51, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22116401

ABSTRACT

The meiosis-specific synaptonemal complex protein SYCP3 has been reported to be aberrantly expressed in tumours. However, in contrast to its well-defined function in meiosis, its possible role in mitotic cells is entirely unknown. Here, we show that SYCP3 is expressed in a range of primary tumours and that it impairs chromosomal integrity in mitotic cells. Expression of SYCP3 inhibits the homologous recombination (HR) pathway mediated by RAD51, inducing hypersensitivity to DNA-damaging agents such as a poly(ADP-ribose) polymerase (PARP) inhibitor and chromosomal instability. SYCP3 forms a complex with BRCA2 and inhibits its role in HR. These findings highlight a new mechanism for chromosomal instability in cancer and extend the range of PARP-inhibitor sensitive tumours to those expressing SYCP3.


Subject(s)
BRCA2 Protein/metabolism , Homologous Recombination , Mitosis/genetics , Nuclear Proteins/metabolism , Aneuploidy , Cell Cycle Proteins , Chromosomal Instability , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , DNA-Binding Proteins , Drug Resistance, Neoplasm/genetics , Gene Silencing , Hep G2 Cells , Humans , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors , Protein Binding , Rad51 Recombinase/metabolism , Radiation Tolerance/genetics , Sister Chromatid Exchange
11.
Genes Cells ; 16(6): 714-27, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21605288

ABSTRACT

Defects in the FANCJ/BRIP1 helicase gene are associated with genome instability disorders such as familial breast cancer or Fanconi anemia (FA). Although FANCJ has an in vitro activity to resolve G-quadruplex (G4) structures, and FANCJ ortholog in C. elegans prevents G4-associated deletions during replication, how FANCJ loss affects genome integrity in higher organisms remains unclear. Here, we report that FANCJ, but not other FA genes FANCD2 or FANCC, protected against large-scale genomic deletion that occurred frequently at the rearranged immunoglobulin heavy chain (IgH) locus in chicken DT40 cell line, suggesting that FancJ protects the genome independently of the FA ubiquitination pathway. In a more unbiased approach using array-comparative genomic hybridization, we identified de novo deletions as well as amplifications in fancj cells kept in culture for 2 months. A cluster of G4 sequence motifs was found near the breakpoint of one amplified region, but G4 sequence motifs were not detected at the breakpoints of two deleted regions. These results collectively suggest that, unlike in C. elegans, actions of vertebrate FANCJ to promote genome stability may not be limited to protection against the G4-mediated gene deletions.


Subject(s)
Fanconi Anemia Complementation Group L Protein/metabolism , Genomic Instability/genetics , RNA Helicases/metabolism , Animals , Base Sequence , Cell Line , Chickens , Comparative Genomic Hybridization , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group L Protein/genetics , G-Quadruplexes , Gene Amplification/genetics , Gene Conversion/genetics , Gene Deletion , Gene Order , Gene Rearrangement/genetics , Gene Targeting , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Molecular Sequence Data , Nucleoside Deaminases/genetics , Nucleoside Deaminases/metabolism , RNA Helicases/genetics , Sequence Alignment
12.
PLoS One ; 5(10): e13554, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-21048951

ABSTRACT

Chromosome translocations induced by DNA damaging agents, such as ionizing radiation and certain chemotherapies, alter genetic information resulting in malignant transformation. Abrogation or loss of the ataxia-telangiectasia mutated (ATM) protein, a DNA damage signaling regulator, increases the incidence of chromosome translocations. However, how ATM protects cells from chromosome translocations is still unclear. Chromosome translocations involving the MLL gene on 11q23 are the most frequent chromosome abnormalities in secondary leukemias associated with chemotherapy employing etoposide, a topoisomerase II poison. Here we show that ATM deficiency results in the excessive binding of the DNA recombination protein RAD51 at the translocation breakpoint hotspot of 11q23 chromosome translocation after etoposide exposure. Binding of Replication protein A (RPA) and the chromatin remodeler INO80, which facilitate RAD51 loading on damaged DNA, to the hotspot were also increased by ATM deficiency. Thus, in addition to activating DNA damage signaling, ATM may avert chromosome translocations by preventing excessive loading of recombinational repair proteins onto translocation breakpoint hotspots.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Recombination, Genetic , Translocation, Genetic , Tumor Suppressor Proteins/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Chromosomes, Human, Pair 11 , DNA-Binding Proteins/genetics , Etoposide/pharmacology , Histone-Lysine N-Methyltransferase , Humans , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Rad51 Recombinase/metabolism , Replication Protein A/metabolism , Tumor Suppressor Proteins/genetics
13.
Nucleic Acids Res ; 37(12): 3959-68, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19403737

ABSTRACT

Rad51C is a central component of two complexes formed by five Rad51 paralogs in vertebrates. These complexes are involved in repairing DNA double-strand breaks through homologous recombination. Despite accumulating evidence suggesting that the paralogs may prevent aneuploidy by controlling centrosome integrity, Rad51C's role in maintaining chromosome stability remains unclear. Here we demonstrate that Rad51C deficiency leads to both centrosome aberrations in an ATR-Chk1-dependent manner and increased aneuploidy in human cells. While it was reported that Rad51C deficiency did not cause centrosome aberrations in interphase in hamster cells, such aberrations were observed in interphase in HCT116 cells with Rad51C dysfunction. Caffeine treatment and down-regulation of ATR, but not that of ATM, reduced the frequency of centrosome aberrations in the mutant cells. Silencing of Rad51C by RNA interference in HT1080 cells resulted in similar aberrations. Treatment with a Chk1 inhibitor and silencing of Chk1 also reduced the frequency in HCT116 mutants. Accumulation of Chk1 at the centrosome and nuclear foci of gamma H2AX were increased in the mutants. Moreover, the mutant cells had a higher frequency of aneuploidy. These findings indicate that the ATR-Chk1 pathway plays a role in increased centrosome aberrations induced by Rad51C dysfunction.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/ultrastructure , DNA-Binding Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Aneuploidy , Ataxia Telangiectasia Mutated Proteins , Cell Line, Tumor , Checkpoint Kinase 1 , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Humans , RNA Interference , Recombination, Genetic
14.
Nat Struct Mol Biol ; 15(11): 1138-46, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18931676

ABSTRACT

In response to DNA damage or replication fork stress, the Fanconi anemia pathway is activated, leading to monoubiquitination of FANCD2 and FANCI and their colocalization in foci. Here we show that, in the chicken DT40 cell system, multiple alanine-substitution mutations in six conserved and clustered Ser/Thr-Gln motifs of FANCI largely abrogate monoubiquitination and focus formation of both FANCI and FANCD2, resulting in loss of DNA repair function. Conversely, FANCI carrying phosphomimic mutations on the same six residues induces constitutive monoubiquitination and focus formation of FANCI and FANCD2, and protects against cell killing and chromosome breakage by DNA interstrand cross-linking agents. We propose that the multiple phosphorylation of FANCI serves as a molecular switch in activation of the Fanconi anemia pathway. Mutational analysis of putative phosphorylation sites in human FANCI indicates that this switch is evolutionarily conserved.


Subject(s)
DNA Damage , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/metabolism , Signal Transduction/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Caffeine/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chickens , DNA Mutational Analysis , DNA Repair , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Humans , Molecular Mimicry , Phosphodiesterase Inhibitors/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ubiquitin/metabolism
15.
Nucleic Acids Res ; 35(19): 6571-87, 2007.
Article in English | MEDLINE | ID: mdl-17905820

ABSTRACT

The RDM1 gene encodes a RNA recognition motif (RRM)-containing protein involved in the cellular response to the anti-cancer drug cisplatin in vertebrates. We previously reported a cDNA encoding the full-length human RDM1 protein. Here, we describe the identification of 11 human cDNAs encoding RDM1 protein isoforms. This repertoire is generated by alternative pre-mRNA splicing and differential usage of two translational start sites, resulting in proteins with long or short N-terminus and a great diversity in the exonic composition of their C-terminus. By using tagged proteins and fluorescent microscopy, we examined the subcellular distribution of full-length RDM1 (renamed RDM1alpha), and other RDM1 isoforms. We show that RDM1alpha undergoes subcellular redistribution and nucleolar accumulation in response to proteotoxic stress and mild heat shock. In unstressed cells, the long N-terminal isoforms displayed distinct subcellular distribution patterns, ranging from a predominantly cytoplasmic to almost exclusive nuclear localization, suggesting functional differences among the RDM1 proteins. However, all isoforms underwent stress-induced nucleolar accumulation. We identified nuclear and nucleolar localization determinants as well as domains conferring cytoplasmic retention to the RDM1 proteins. Finally, RDM1 null chicken DT40 cells displayed an increased sensitivity to heat shock, compared to wild-type (wt) cells, suggesting a function for RDM1 in the heat-shock response.


Subject(s)
Cell Nucleolus/chemistry , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Heat-Shock Response , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Alternative Splicing , Animals , Base Sequence , Cell Line , Cell Nucleus/chemistry , Chickens , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/chemistry , DNA-Binding Proteins/chemistry , Dactinomycin/pharmacology , Exons , Gene Deletion , Humans , Molecular Sequence Data , Nucleic Acid Synthesis Inhibitors/pharmacology , Proteasome Inhibitors , Protein Isoforms/analysis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
16.
Cancer Res ; 66(12): 6018-24, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778173

ABSTRACT

The Rad51-like proteins, Rad51B, Rad51C, Rad51D, XRCC2, and XRCC3, have been shown to form two distinct complexes and seem to assist Rad51 in the early stages of homologous recombination. Although these proteins share sequence similarity with Rad51, they do not show functional redundancy. Among them, Rad51B is unique in that the gene maps to the human chromosome 14q23-24, the region frequently involved in balanced chromosome translocations in benign tumors particularly in uterine leiomyomas. Despite accumulating descriptive evidence of altered Rad51B function in these tumors, the biological significance of this aberration is still unknown. To assess the significance of reduced Rad51B function, we deleted the gene in the human colon cancer cell line HCT116 by gene targeting. Here, we show that haploinsufficiency of RAD51B causes mild hypersensitivity to DNA-damaging agents, a mild reduction in sister chromatid exchange, impaired Rad51 focus formation, and an increase in chromosome aberrations. Remarkably, haploinsufficiency of RAD51B leads to centrosome fragmentation and aneuploidy. In addition, an approximately 50% reduction in RAD51B mRNA levels by RNA interference also leads to centrosome fragmentation in the human fibrosarcoma cell line HT1080. These findings suggest that the proper biallelic expression of RAD51B is required for the maintenance of chromosome integrity in human cells.


Subject(s)
Aneuploidy , Centrosome/metabolism , Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Chromosome Aberrations , Colonic Neoplasms/metabolism , DNA Damage , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Gene Targeting , HCT116 Cells , Haploidy , Humans , RNA, Small Interfering/genetics , Sister Chromatid Exchange
17.
Nucleic Acids Res ; 34(3): 880-92, 2006.
Article in English | MEDLINE | ID: mdl-16456034

ABSTRACT

The Mus81-Eme1 complex is a structure-specific endonuclease that preferentially cleaves nicked Holliday junctions, 3'-flap structures and aberrant replication fork structures. Mus81-/- mice have been shown to exhibit spontaneous chromosomal aberrations and, in one of two models, a predisposition to cancers. The molecular mechanisms underlying its role in chromosome integrity, however, are largely unknown. To clarify the role of Mus81 in human cells, we deleted the gene in the human colon cancer cell line HCT116 by gene targeting. Here we demonstrate that Mus81 confers resistance to DNA crosslinking agents and slight resistance to other DNA-damaging agents. Mus81 deficiency spontaneously promotes chromosome damage such as breaks and activates the intra-S-phase checkpoint through the ATM-Chk1/Chk2 pathways. Furthermore, Mus81 deficiency activates the G2/M checkpoint through the ATM-Chk2 pathway and promotes DNA rereplication. Increased rereplication is reversed by the ectopic expression of Cdk1. Haploinsufficiency of Mus81 or Eme1 also causes similar phenotypes. These findings suggest that a complex network of the checkpoint pathways that respond to DNA double-strand breaks may participate in some of the phenotypes associated with Mus81 or Eme1 deficiency.


Subject(s)
Cell Cycle , Chromosomal Instability , DNA-Binding Proteins/physiology , Endodeoxyribonucleases/physiology , Endonucleases/physiology , Polyploidy , CDC2 Protein Kinase/metabolism , Cell Line , DNA Damage , DNA Replication , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/genetics , Endonucleases/genetics , G2 Phase , Gene Deletion , Gene Targeting , Heterozygote , Humans , Mitosis , S Phase
18.
EMBO J ; 23(3): 670-80, 2004 Feb 11.
Article in English | MEDLINE | ID: mdl-14749735

ABSTRACT

XRCC3 was inactivated in human cells by gene targeting. Consistent with its role in homologous recombination, XRCC3(-/-) cells showed a two-fold sensitivity to DNA cross-linking agents, a mild reduction in sister chromatid exchange, impaired Rad51 focus formation and elevated chromosome aberrations. Furthermore, endoreduplication was increased five- seven-fold in the mutants. The T241M variant of XRCC3 has been associated with an increased cancer risk. Expression of the wild-type cDNA restored this phenotype, while expression of the variant restored the defective recombinational repair, but not the increased endoreduplication. RPA, a protein essential for homologous recombination and DNA replication, is associated with XRCC3 and Rad52. Overexpression of RPA promoted endoreduplication, which was partially complemented by overexpression of the wild-type XRCC3 protein, but not by overexpression of the variant protein. Overexpression of Rad52 prevented endoreduplication in RPA-overexpressing cells, in XRCC3(-/-) cells and in the variant-expressing cells, suggesting that deregulated RPA was responsible for the increased endoreduplication. These observations offer the first genetic evidence for the association between homologous recombination and replication initiation having a role in cancer susceptibility.


Subject(s)
Agmatine/analogs & derivatives , DNA Repair/physiology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Ploidies , Recombination, Genetic , Agmatine/pharmacology , Cell Line , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Cross-Linking Reagents/pharmacology , DNA Damage/drug effects , DNA Repair/genetics , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Gene Deletion , Gene Expression , Gene Targeting , Humans , Mitomycin/pharmacology , Rad52 DNA Repair and Recombination Protein , Replication Protein A , Succinates/pharmacology
19.
EMBO J ; 21(1-2): 175-80, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11782437

ABSTRACT

In human somatic cells, homologous recombination is a rare event. To facilitate the targeted modification of the genome for research and gene therapy applications, efforts should be directed toward understanding the molecular mechanisms of homologous recombination in human cells. Although human genes homologous to members of the RAD52 epistasis group in yeast have been identified, no genes have been demonstrated to play a role in homologous recombination in human cells. Here, we report that RAD54B plays a critical role in targeted integration in human cells. Inactivation of RAD54B in a colon cancer cell line resulted in severe reduction of targeted integration frequency. Sensitivity to DNA-damaging agents and sister-chromatid exchange were not affected in RAD54B-deficient cells. Parts of these phenotypes were similar to those of Saccharomyces cerevisiae tid1/rdh54 mutants, suggesting that RAD54B may be a human homolog of TID1/RDH54. In yeast, TID1/RDH54 acts in the recombinational repair pathway via roles partially overlapping those of RAD54. Our findings provide the first genetic evidence that the mitotic recombination pathway is functionally conserved from yeast to humans.


Subject(s)
Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Recombination, Genetic , Base Sequence , Cell Division/genetics , Cell Survival/genetics , Colonic Neoplasms/genetics , DNA Helicases , DNA Repair Enzymes , DNA Topoisomerases , DNA, Neoplasm/genetics , Fungal Proteins/genetics , Gene Targeting , Genes, Fungal , Humans , Phenotype , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sister Chromatid Exchange , Species Specificity , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...