Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters








Publication year range
1.
Biol Pharm Bull ; 47(7): 1338-1344, 2024.
Article in English | MEDLINE | ID: mdl-39048355

ABSTRACT

The initiation of DNA replication is tightly controlled by the licensing system that loads replicative DNA helicases onto replication origins to form pre-replicative complexes (pre-RCs) once per cell cycle. Cdc10-dependent transcript 1 (Cdt1) plays an essential role in the licensing reaction by recruiting mini-chromosome maintenance (MCM) complexes, which are eukaryotic replicative DNA helicases, to their origins via direct protein-protein interactions. Cdt1 interacts with other pre-RC components, the origin recognition complex, and the cell division cycle 6 (Cdc6) protein; however, the molecular mechanism by which Cdt1 functions in the MCM complex loading process has not been fully elucidated. Here, we analyzed the protein-protein interactions of recombinant Cdt1 and observed that Cdt1 self-associates via the central region of the molecule, which is inhibited by the endogenous licensing inhibitor, geminin. Mutation of two ß-strands of the winged-helix domain in the central region of Cdt1 attenuated its self-association but could still interact with other pre-RC components and DNA similarly to wild-type Cdt1. Moreover, the Cdt1 mutant showed decreased licensing activity in Xenopus egg extracts. Together, these results suggest that the self-association of Cdt1 is crucial for licensing.


Subject(s)
Cell Cycle Proteins , Geminin , Animals , Geminin/metabolism , Geminin/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA Replication , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Xenopus laevis , Protein Domains , Xenopus , Humans , DNA-Binding Proteins
2.
Biochem Biophys Rep ; 29: 101222, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35146135

ABSTRACT

Calreticulin (CRT), a chaperone typically located in the endoplasmic reticulum (ER), is known to translocate to the cell surface in response to anticancer drugs. Cell surface CRT (ecto-CRT) on apoptotic or pre-apoptotic cells serves as an "eat me" signal that can promote phagocytosis. In this study, we observed the biphasic (early transient and late sustained) increase of ecto-CRT on HT-29 cells after treatment with oxaliplatin (L-OHP). To investigate the role of ecto-CRT that accumulates in the early and late phases as "eat me" signals, we examined the phagocytosis of HT-29 cells by macrophage-like cells and dendritic cell (DC) -like cells prepared from THP-1 cells. The results indicated that the early ecto-CRT-expressed cells were phagocytosed by immature DC-like cells, and the late ecto-CRT-expressed cells were phagocytosed primarily by macrophage-like cells, while mature DC-like cells did not respond to the either class of ecto-CRT-expressed cells. Both types of phagocytotic events were inhibited by CRT Blocking Peptide, suggesting that such events depended on the ecto-CRT. Our results suggested that the early increase of ecto-CRT is related to phagocytosis as part of immunogenic cell death (ICD), while the late increase of ecto-CRT is related to the removal of apoptotic cells by macrophages.

3.
Gene ; 787: 145647, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-33845136

ABSTRACT

RecQ4, a member of the RecQ helicase family, is required for the maintenance of genome integrity. RecQ4 has been shown to promote the following two DNA double-strand break (DSB) repair pathways: non-homologous end joining (NHEJ) and homologous recombination (HR). However, its molecular function has not been fully elucidated. In the present study, we aimed to investigate the role of RecQ4 in NHEJ using Xenopus egg extracts. The N-terminal 598 amino acid region of Xenopus RecQ4 (N598), which lacks a central helicase domain and a downstream C-terminal region, was added to the extracts and its effect on the joining of DNA ends was analyzed. We found that N598 inhibited the joining of linearized DNA ends in the extracts. In addition, N598 inhibited DSB-induced chromatin binding of Ku70, which is essential for NHEJ, while the DSB-induced chromatin binding of the HR-associated proteins, replication protein A (RPA) and Rad51, increased upon the addition of N598. These results suggest that RecQ4 possibly influences the choice of the DSB repair pathway by influencing the association of the Ku heterodimer with the DNA ends.


Subject(s)
DNA End-Joining Repair , Ku Autoantigen/metabolism , RecQ Helicases/metabolism , Xenopus Proteins/physiology , Animals , Chromatin , DNA/metabolism , Ku Autoantigen/antagonists & inhibitors , Protein Binding , RecQ Helicases/genetics , Xenopus laevis
4.
Biol Pharm Bull ; 43(10): 1595-1599, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32727970

ABSTRACT

Calreticulin (CRT) and calnexin (CNX), homologous major chaperones in the endoplasmic reticulum (ER), are known to translocate to the cell surface in response to chemotherapeutic agents, such as mitoxantrone (MIT), and cellular stresses, including apoptosis. Cell surface CRT (ecto-CRT) is relevant to the phagocytic uptake of cancer cells and dying cells, and pre-apoptotic exposure of CRT has been reported to result in enhanced immunogenicity of dying tumor cells, serving as a damage-associated molecular pattern (DAMP). In this study, HT-29 cells were treated with MIT to induce ER stress, and ecto-CRT and cell surface CNX were quantified by flow cytometry in the absence or presence of caspase inhibitors, a calpain inhibitor, or a scavenger of reactive oxygen species. The biphasic (early transient and late sustained) increase of ecto-CRT on HT-29 cells was observed after treatment with MIT. We confirmed that the early increase in ecto-CRT after 4 h of MIT treatment was not related to apoptosis, whereas the increase of ecto-CRT, as well as that of cell-surface CNX, during the later stage of treatment was caspase dependent and related to apoptosis. In addition, our results suggested that the early peak of ecto-CRT was mediated by activation of caspase 8 by ER stress. Thus, the physiological significance of the late increases in cell-surface CRT and/or CNX might be considered an "eat-me signal" for the removal of dead cells by phagocytosis, while the early increase in ecto-CRT caused by ER stress might enhance the immunogenicity of stressed tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Calreticulin/metabolism , Cell Membrane/metabolism , Mitoxantrone/pharmacology , Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , Calnexin/analysis , Calnexin/metabolism , Calreticulin/analysis , Cell Membrane/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , HT29 Cells , Humans , Mitoxantrone/therapeutic use , Neoplasms/immunology , Neoplasms/pathology , Phagocytosis/drug effects , Reactive Oxygen Species/metabolism
5.
Biochem Biophys Res Commun ; 490(4): 1375-1380, 2017 09 02.
Article in English | MEDLINE | ID: mdl-28694193

ABSTRACT

The initiation of DNA replication is strictly regulated by multiple mechanisms to ensure precise duplication of chromosomes. In higher eukaryotes, activity of the Cdt1 protein is temporally regulated during the cell cycle, and deregulation of Cdt1 induces DNA re-replication. In previous studies, we showed that excess Cdt1 inhibits DNA replication by suppressing progression of replication forks in Xenopus egg extracts. Here, we investigated the functional regions of Cdt1 that are required for the inhibition of DNA replication. We constructed a series of N-terminally or C-terminally deleted mutants of Cdt1 and examined their inhibitory effects on DNA replication in Xenopus egg extracts. Our results showed that the region spanning amino acids (a. a.) 255-620 is required for efficient inhibition of DNA replication, and that, within this region, a. a. 255-289 have a critical role in inhibition. Moreover, one of the Cdt1 mutants, Cdt1 R285A, was compromised with respect to the licensing activity but still inhibited DNA replication. This result suggests that Cdt1 has an unforeseen function in the negative regulation of DNA replication, and that this function is located within a molecular region that is distinct from those required for the licensing activity.


Subject(s)
Cell Cycle Proteins/genetics , Chromatin/chemistry , DNA Replication , DNA-Binding Proteins/genetics , Geminin/genetics , Ovum/chemistry , Xenopus Proteins/genetics , Xenopus laevis/genetics , Animals , Binding Sites , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Chromatin/metabolism , Cloning, Molecular , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Geminin/metabolism , Gene Expression , Male , Mutation , Ovum/cytology , Ovum/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spermatozoa/chemistry , Spermatozoa/cytology , Spermatozoa/metabolism , Structure-Activity Relationship , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
6.
Biochem Biophys Res Commun ; 470(2): 405-410, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26773501

ABSTRACT

Cdt1 is a protein essential for initiation of DNA replication; it recruits MCM helicase, a core component of the replicative DNA helicase, onto replication origins. In our previous study, we showed that addition of excess Cdt1 inhibits nascent strand elongation during DNA replication in Xenopus egg extracts. In the present study, we investigated the mechanism behind the inhibitory effect of Cdt1. We found that addition of recombinant Cdt1 inhibited nascent DNA synthesis in a reinitiation-independent manner. To identify the mechanism by which Cdt1 inhibits nascent strand elongation, the effect of Cdt1 on loading of Mcm4 and Rpa70 onto chromatin was examined. The results showed that Cdt1 suppressed the excessive Rpa70 binding caused by extensive, aphidicolin-induced DNA unwinding; this unwinding occurs between stalled DNA polymerases and advancing replication forks. These findings suggested that excess Cdt1 suppressed the progression of replication forks.


Subject(s)
Cell Cycle Proteins/genetics , DNA Replication/genetics , Ovum/physiology , Transcription Elongation, Genetic/physiology , Animals , Xenopus laevis
7.
Biochem Biophys Res Commun ; 452(1): 48-52, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25139235

ABSTRACT

WRNIP1 (WRN-interacting protein 1) was first identified as a factor that interacts with WRN, the protein that is defective in Werner syndrome (WS). WRNIP1 associates with DNA polymerase η (Polη), but the biological significance of this interaction remains unknown. In this study, we analyzed the functional interaction between WRNIP1 and Polη by generating knockouts of both genes in DT40 chicken cells. Disruption of WRNIP1 in Polη-disrupted (POLH(-/-)) cells suppressed the phenotypes associated with the loss of Polη: sensitivity to ultraviolet light (UV), delayed repair of cyclobutane pyrimidine dimers (CPD), elevated frequency of mutation, elevated levels of UV-induced sister chromatid exchange (SCE), and reduced rate of fork progression after UV irradiation. These results suggest that WRNIP1 functions upstream of Polη in the response to UV irradiation.


Subject(s)
Carrier Proteins/physiology , DNA Damage , DNA-Binding Proteins/physiology , DNA-Directed DNA Polymerase/metabolism , DNA/radiation effects , Ultraviolet Rays , ATPases Associated with Diverse Cellular Activities , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Sister Chromatid Exchange
8.
J Biol Chem ; 289(16): 11374-11384, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24573676

ABSTRACT

The replication fork temporarily stalls when encountering an obstacle on the DNA, and replication resumes after the barrier is removed. Simultaneously, activation of the replication checkpoint delays the progression of S phase and inhibits late origin firing. Camptothecin (CPT), a topoisomerase I (Top1) inhibitor, acts as a DNA replication barrier by inducing the covalent retention of Top1 on DNA. The Timeless-Tipin complex, a component of the replication fork machinery, plays a role in replication checkpoint activation and stabilization of the replication fork. However, the role of the Timeless-Tipin complex in overcoming the CPT-induced replication block remains elusive. Here, we generated viable TIPIN gene knock-out (KO) DT40 cells showing delayed S phase progression and increased cell death. TIPIN KO cells were hypersensitive to CPT. However, homologous recombination and replication checkpoint were activated normally, whereas DNA synthesis activity was markedly decreased in CPT-treated TIPIN KO cells. Proteasome-dependent degradation of chromatin-bound Top1 was induced in TIPIN KO cells upon CPT treatment, and pretreatment with aphidicolin, a DNA polymerase inhibitor, suppressed both CPT sensitivity and Top1 degradation. Taken together, our data indicate that replication forks formed without Tipin may collide at a high rate with Top1 retained on DNA by CPT treatment, leading to CPT hypersensitivity and Top1 degradation in TIPIN KO cells.


Subject(s)
Avian Proteins/metabolism , Camptothecin/pharmacology , DNA Replication/drug effects , DNA Topoisomerases, Type I/metabolism , Nuclear Proteins/metabolism , Topoisomerase I Inhibitors/pharmacology , Animals , Avian Proteins/genetics , Cell Death/drug effects , Cell Death/genetics , Cell Line , Chickens , DNA/biosynthesis , DNA/genetics , DNA Replication/physiology , DNA Topoisomerases, Type I/genetics , Gene Knockdown Techniques , Nuclear Proteins/genetics , Proteolysis/drug effects , S Phase/drug effects , S Phase/physiology
9.
Biochem Biophys Res Commun ; 427(3): 682-6, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23036200

ABSTRACT

Saccharomyces cerevisiae RecQ helicase (Sgs1) combines with DNA topoisomerase III (Top3) and RecQ-mediated genome instability 1 (Rmi1) to form an evolutionarily conserved complex that is required for processing homologous recombination intermediates and restarting collapsed replication forks. It was previously reported that Rmi1 contributes to sister chromatid cohesion; however, the underlying molecular mechanism has been unclear. In the present study, Rmi1 was found to be enriched at the region close to an early-firing replication origin when replication forks were arrested near their origins in the presence of hydroxyurea. Genetic analyses revealed that Rmi1 promoted sister chromatid cohesion in a process that was distinct from both the cohesion establishment pathway involving Ctf4, Csm3, and Chl1 and the pathway involving the acetylation of Smc3. On the other hand, Rmi1 seemed to function in a pathway involving the Ctf18-RFC complex and Mrc1, which were previously predicted to regulate leading-strand DNA replication.


Subject(s)
DNA-Binding Proteins/metabolism , Homologous Recombination , Replication Protein C/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Acetylation , Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication , DNA-Binding Proteins/genetics , Nuclear Proteins/metabolism , Protein Conformation , RecQ Helicases/metabolism , S Phase , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
10.
Genes Cells ; 16(10): 1050-62, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21895891

ABSTRACT

The nucleosome, which is composed of DNA wrapped around a histone octamer, is a fundamental unit of chromatin and is duplicated during the eukaryotic DNA replication process. The evolutionarily conserved histone chaperone cell cycle gene 1 (CCG1) interacting factor A/anti-silencing function 1 (CIA/Asf1) is involved in histone transfer and nucleosome reassembly during DNA replication. CIA/Asf1 has been reported to split the histone (H3-H4)(2) tetramer into histone H3-H4 dimer(s) in vitro, raising a possibility that, in DNA replication, CIA/Asf1 is involved in nucleosome disassembly and the promotion of semi-conservative histone H3-H4 dimer deposition onto each daughter strand in vivo. Despite numerous studies on the functional roles of CIA/Asf1, its mechanistic role(s) remains elusive because of lack of biochemical analyses. The biochemical studies described here show that a V94R CIA/Asf1 mutant, which lacks histone (H3-H4)(2) tetramer splitting activity, does not form efficiently a quaternary complex with histones H3-H4 and the minichromosome maintenance 2 (Mcm2) subunit of the Mcm2-7 replicative DNA helicase. Interestingly, the mutant enhances nascent DNA strand synthesis in a cell-free chromosomal DNA replication system using Xenopus egg extracts. These results suggest that CIA/Asf1 in the CIA/Asf1-H3-H4-Mcm2 complex, which is considered to be an intermediate in histone transfer during DNA replication, negatively regulates the progression of the replication fork.


Subject(s)
DNA Replication/physiology , Histone Chaperones/metabolism , Nucleosomes/metabolism , Animals , Chromatin Assembly and Disassembly , Histone Chaperones/genetics , Histones/metabolism , Kinetics , Models, Molecular , Mutation/genetics , Protein Binding , Xenopus
11.
Biochem Biophys Res Commun ; 414(2): 298-303, 2011 Oct 22.
Article in English | MEDLINE | ID: mdl-21945441

ABSTRACT

Claspin was originally identified as a Check1 (Chk1)-interacting protein. Claspin and Rad17 are reportedly involved in the DNA damage-induced phosphorylation of Chk1, a hallmark of checkpoint activation. To understand the cellular functions of Claspin and the functional relationship between Claspin and Rad17, we generated Claspin(-/-) and Claspin(-/-)/RAD17(-) cells using chicken DT40 cells, which contain an exogenously introduced Claspin that can be suppressed by the addition of doxycycline (Dox). In the presence of Dox, Claspin(-/-) cells ceased growth within 2 days, leading to cell death. In addition, a remarkable reduction in the rate of DNA elongation was observed in Claspin-depleted cells, suggesting that Claspin plays a critical role in DNA replication in the absence of exogenous stress. When cells were exposed to methyl methanesulfonate (MMS), a DNA damaging agent, RAD17(-) cells showed a greater defect in checkpoint activation than Claspin(-/-) cells as monitored by progression of cell cycle and phosphorylation of Chk1. Knocking out RAD17 gene showed almost no additive effects on cell death and DNA elongation rates in Claspin-depleted cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cell Line , Chickens , DNA Replication/genetics , Gene Knockout Techniques , Humans
12.
Biol Pharm Bull ; 34(8): 1314-8, 2011.
Article in English | MEDLINE | ID: mdl-21804224

ABSTRACT

Werner interacting protein 1 (WRNIP1) that is highly conserved from Escherichia coli to human was originally identified as a protein that interacts with the Werner syndrome responsible gene product (WRN). Here, human WRNIP1 and WRN are shown to bind to template-primer DNA, and WRNIP1, but not WRN, requires ATP for DNA binding. Under conditions of a limiting amount of WRN, WRNIP1 facilitated binding of WRN to DNA in a dose-dependent manner. However, WRNIP1 did not stimulate the DNA helicase activity of WRN, and WRN displaced pre-bound WRNIP1 from DNA. Functional relationships between WRNIP1 and WRN will be discussed.


Subject(s)
Carrier Proteins/metabolism , DNA Primers/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Exodeoxyribonucleases/metabolism , RecQ Helicases/metabolism , Werner Syndrome/genetics , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphate/metabolism , Humans , Protein Binding , Werner Syndrome/metabolism , Werner Syndrome Helicase
13.
J Biol Chem ; 286(35): 30504-30512, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21757688

ABSTRACT

Ordered nucleosome disassembly and reassembly are required for eukaryotic DNA replication. The facilitates chromatin transcription (FACT) complex, a histone chaperone comprising Spt16 and SSRP1, is involved in DNA replication as well as transcription. FACT associates with the MCM helicase, which is involved in DNA replication initiation and elongation. Although the FACT-MCM complex is reported to regulate DNA replication initiation, its functional role in DNA replication elongation remains elusive. To elucidate the functional role of FACT in replication fork progression during DNA elongation in the cells, we generated and analyzed conditional SSRP1 gene knock-out chicken (Gallus gallus) DT40 cells. SSRP1-depleted cells ceased to grow and exhibited a delay in S-phase cell cycle progression, although SSRP1 depletion did not affect the level of chromatin-bound DNA polymerase α or nucleosome reassembly on daughter strands. The tracking length of newly synthesized DNA, but not origin firing, was reduced in SSRP1-depleted cells, suggesting that the S-phase cell cycle delay is mainly due to the inhibition of replication fork progression rather than to defects in the initiation of DNA replication in these cells. We discuss the mechanisms of how FACT promotes replication fork progression in the cells.


Subject(s)
Chromatin/chemistry , DNA Replication , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Histone Chaperones/chemistry , Transcription, Genetic , Transcriptional Elongation Factors/metabolism , Animals , Cell Cycle , Chickens , Epigenesis, Genetic , Flow Cytometry/methods , Histones/chemistry , Humans , Molecular Chaperones/metabolism , Nucleosomes/metabolism , S Phase
14.
Biochim Biophys Acta ; 1813(6): 1129-36, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21232560

ABSTRACT

The facilitates chromatin transcription (FACT) complex affects nuclear DNA transactions in a chromatin context. Though the involvement of FACT in eukaryotic DNA replication has been revealed, a clear understanding of its biochemical behavior during DNA replication still remains elusive. Here, we analyzed the chromatin-binding dynamics of FACT using Xenopus egg extract cell-free system. We found that FACT has at least two distinct chromatin-binding phases: (1) a rapid chromatin-binding phase at the onset of DNA replication that did not involve origin licensing and (2) a second phase of chromatin binding that initiated after origin licensing. Intriguingly, early-binding FACT dissociated from chromatin when DNA replication was blocked by the addition of Cdc6 in the licensed state before origin firing. Cdc6-induced removal of FACT was blocked by the inhibition of origin licensing with geminin, but not by suppressing the activity of DNA polymerases, CDK, or Cdc7. Furthermore, chromatin transfer experiments revealed that impairing the later binding of FACT severely compromises DNA replication activity. Taken together, we propose that even though FACT has rapid chromatin-binding activity, the binding pattern of FACT on chromatin changes after origin licensing, which may contribute to the establishment of its functional link to the DNA replication machinery.


Subject(s)
Chromatin/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Transcriptional Elongation Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/genetics , DNA-Binding Proteins/genetics , Eukaryotic Cells/metabolism , Female , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , High Mobility Group Proteins/genetics , Histone Chaperones/genetics , Histone Chaperones/metabolism , Humans , Immunoblotting , Kinetics , Male , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oocytes/metabolism , Protein Binding , Spermatozoa/metabolism , Time Factors , Transcriptional Elongation Factors/genetics , Xenopus laevis
15.
Biochim Biophys Acta ; 1813(3): 473-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21256165

ABSTRACT

Rothmund-Thomson syndrome (RTS) is a rare genetic disorder characterized by premature aging, developmental abnormalities, and a predisposition to cancer. RTS is caused by mutations in the RECQL4 gene, which encodes one of the five human RecQ helicases. To identify the cellular functions of RECQL4, we generated a chicken DT40 cell line in which RECQL4 expression could be turned off by doxycycline (Dox). Upon exposure to Dox, cells stopped growing and underwent apoptosis. The cells could be rescued by expression of the N-terminal region of RECQL4 (amino acids 1-496), which lacks the helicase domain and has sequence similarity to yeast Sld2, which plays an essential function in the initiation of DNA replication in Saccharomyces cerevisiae. Smaller fragments of the N-terminal region of RECQL4 did not rescue the cells from lethality. RECQL4 gene knockout cells complemented with RECQL4 (1-496) showed relatively high sensitivity to DNA damaging agents that induce double strand breaks and cross-links, suggesting that the C-terminal region including the helicase domain of RECQL4 is involved in the repair of certain types of DNA lesions.


Subject(s)
Cell Survival , RecQ Helicases/genetics , RecQ Helicases/metabolism , Rothmund-Thomson Syndrome/genetics , Animals , Anti-Bacterial Agents/pharmacology , Cell Death , Cell Line , Chickens , DNA Helicases/metabolism , DNA Repair , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Humans , Protein Structure, Tertiary , RecQ Helicases/chemistry , Rothmund-Thomson Syndrome/metabolism
16.
J Nucleic Acids ; 20102010 Aug 05.
Article in English | MEDLINE | ID: mdl-20811569

ABSTRACT

Reactive oxygen species (ROSs) are produced during normal cellular metabolism, particularly by respiration in mitochondria, and these ROSs are considered to cause oxidative damage to macromolecules, including DNA. In our previous paper, we found no indication that depletion of mitochondrial superoxide dismutase, SOD2, resulted in an increase in DNA damage. In this paper, we examined SOD1, which is distributed in the cytoplasm, nucleus, and mitochondrial intermembrane space. We generated conditional SOD1 knockout cells from chicken DT40 cells and analyzed their phenotypes. The results revealed that SOD1 was essential for viability and that depletion of SOD1, especially nuclear SOD1, increased sister chromatid exchange (SCE) frequency, suggesting that superoxide is generated in or near the nucleus and that nuclear SOD1 functions as a guardian of the genome. Furthermore, we found that ascorbic acid could offset the defects caused by SOD1 depletion, including cell lethality and increases in SCE frequency and apurinic/apyrimidinic sites.

17.
Nucleic Acids Res ; 38(16): 5409-18, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20421204

ABSTRACT

Mcm2-7 is recruited to eukaryotic origins of DNA replication by origin recognition complex, Cdc6 and Cdt1 thereby licensing the origins. Cdc6 is essential for origin licensing during DNA replication and is readily destabilized from chromatin after Mcm2-7 loading. Here, we show that after origin licensing, deregulation of Cdc6 suppresses DNA replication in Xenopus egg extracts without the involvement of ATM/ATR-dependent checkpoint pathways. DNA replication is arrested specifically after chromatin binding of Cdc7, but before Cdk2-dependent pathways and deregulating Cdc6 after this step does not impair activation of origin firing or elongation. Detailed analyses revealed that Cdc6 deregulation leads to strong suppression of Cdc7-mediated hyperphosphorylation of Mcm4 and subsequent chromatin loading of Cdc45, Sld5 and DNA polymerase α. Mcm2 phosphorylation is also repressed although to a lesser extent. Remarkably, Cdc6 itself does not directly inhibit Cdc7 kinase activity towards Mcm2-4-6-7 in purified systems, rather modulates Mcm2-7 phosphorylation on chromatin context. Taken together, we propose that Cdc6 on chromatin acts as a modulator of Cdc7-mediated phosphorylation of Mcm2-7, and thus destabilization of Cdc6 from chromatin after licensing is a key event ensuring proper transition to the initiation of DNA replication.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA Replication , Protein Serine-Threonine Kinases/antagonists & inhibitors , Xenopus Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Chromatin/enzymology , Cyclin-Dependent Kinase 2/metabolism , DNA-Binding Proteins/metabolism , Humans , Mice , Minichromosome Maintenance Complex Component 2 , Minichromosome Maintenance Complex Component 4 , Minichromosome Maintenance Complex Component 6 , Minichromosome Maintenance Complex Component 7 , Nuclear Proteins/metabolism , Origin Recognition Complex/metabolism , Ovum/enzymology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Xenopus laevis
18.
Protein Sci ; 18(11): 2252-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19722278

ABSTRACT

In eukaryotic replication licensing, Cdt1 plays a key role by recruiting the MCM2-7 complex onto the origin of chromosome. The C-terminal domain of mouse Cdt1 (mCdt1C), the most conserved region in Cdt1, is essential for licensing and directly interacts with the MCM2-7 complex. We have determined the structures of mCdt1CS (mCdt1C_small; residues 452 to 557) and mCdt1CL (mCdt1C_large; residues 420 to 557) using X-ray crystallography and solution NMR spectroscopy, respectively. While the N-terminal 31 residues of mCdt1CL form a flexible loop with a short helix near the middle, the rest of mCdt1C folds into a winged helix structure. Together with the middle domain of mouse Cdt1 (mCdt1M, residues 172-368), this study reveals that Cdt1 is formed with a tandem repeat of the winged helix domain. The winged helix fold is also conserved in other licensing factors including archaeal ORC and Cdc6, which supports an idea that these replication initiators may have evolved from a common ancestor. Based on the structure of mCdt1C, in conjunction with the biochemical analysis, we propose a binding site for the MCM complex within the mCdt1C.


Subject(s)
Binding Sites/genetics , Cell Cycle Proteins/chemistry , Conserved Sequence/genetics , DNA-Binding Proteins/chemistry , Protein Structure, Tertiary/genetics , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Mice , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Sequence Alignment
19.
Genes Genet Syst ; 84(2): 171-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19556710

ABSTRACT

WRN interacting protein 1 (WRNIP1) was originally identified as a protein that interacts with the Werner syndrome responsible gene product (WRN). WRNIP1 is a highly conserved protein from E. coli to humans. Genetic studies in budding yeast suggested that the yeast orthlog of WRNIP1, Mgs1, may function in a DNA damage tolerance pathway that is similar to, but distinct from, the template-switch damage avoidance pathway involving Rad6, Rad18, Rad5, Mms2, and Ubc13. Here we report that human WRNIP1 binds in an ATP dependent manner to both forked DNA that mimics stalled replication forks and to template/primer DNA. We found that WRNIP1 interacts physically with RAD18 and interferes with the binding of RAD18 to forked DNA and to template/primer DNA. In contrast, RAD18 enhances the binding of WRNIP1 to these DNAs, suggesting that WRNIP1 targets DNA bound by RAD18.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Protein Interaction Mapping/methods , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphate/metabolism , Animals , Carrier Proteins/genetics , Cell Line , DNA-Binding Proteins/genetics , Humans , Immunoprecipitation , Nucleic Acid Conformation , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oligonucleotides/metabolism , Protein Binding , Spodoptera , Transfection , Ubiquitin-Protein Ligases
20.
DNA Repair (Amst) ; 8(6): 690-6, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19261547

ABSTRACT

Checkpoints are cellular surveillance and signaling pathways that regulate responses to DNA damage and perturbations of DNA replication. Here we show that high levels of sumoylated Rad52 are present in the mec1 sml1 and rad53 sml1 checkpoint mutants exposed to DNA-damaging agents such as methyl methanesulfonate (MMS) or the DNA replication inhibitor hydroxyurea (HU). The kinase-defective mutant rad53-K227A also showed high levels of Rad52 sumoylation. Elevated levels of Rad52 sumoylation occur in checkpoint mutants proceeding S phase being exposed DNA-damaging agent. Interestingly, chromatin immunoprecipitation (ChIP) on chip analyses revealed non-canonical chromosomal localization of Rad52 in the HU-treated rad53-K227A cells arrested in early S phase: Rad52 localization at dormant and early DNA replication origins. However, such unusual localization was not dependent on the sumoylation of Rad52. In addition, we also found that Rad52 could be highly sumoylated in the absence of Rad51. Double mutation of RAD51 and RAD53 exhibited the similar levels of Rad52 sumoylation to RAD53 single mutation. The significance and regulation mechanism of Rad52 sumoylation by checkpoint pathways will be discussed.


Subject(s)
DNA Replication , DNA, Bacterial/genetics , Mutation/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2 , Chromatin Immunoprecipitation , DNA, Fungal , Flow Cytometry , Immunoblotting , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL