Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Nature ; 567(7747): 267-272, 2019 03.
Article in English | MEDLINE | ID: mdl-30842657

ABSTRACT

Cells often use multiple pathways to repair the same DNA lesion, and the choice of pathway has substantial implications for the fidelity of genome maintenance. DNA interstrand crosslinks covalently link the two strands of DNA, and thereby block replication and transcription; the cytotoxicity of these crosslinks is exploited for chemotherapy. In Xenopus egg extracts, the collision of replication forks with interstrand crosslinks initiates two distinct repair pathways. NEIL3 glycosylase can cleave the crosslink1; however, if this fails, Fanconi anaemia proteins incise the phosphodiester backbone that surrounds the interstrand crosslink, generating a double-strand-break intermediate that is repaired by homologous recombination2. It is not known how the simpler NEIL3 pathway is prioritized over the Fanconi anaemia pathway, which can cause genomic rearrangements. Here we show that the E3 ubiquitin ligase TRAIP is required for both pathways. When two replisomes converge at an interstrand crosslink, TRAIP ubiquitylates the replicative DNA helicase CMG (the complex of CDC45, MCM2-7 and GINS). Short ubiquitin chains recruit NEIL3 through direct binding, whereas longer chains are required for the unloading of CMG by the p97 ATPase, which enables the Fanconi anaemia pathway. Thus, TRAIP controls the choice between the two known pathways of replication-coupled interstrand-crosslink repair. These results, together with our other recent findings3,4 establish TRAIP as a master regulator of CMG unloading and the response of the replisome to obstacles.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Repair , DNA/chemistry , DNA/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA/biosynthesis , DNA Replication , Female , Humans , Minichromosome Maintenance Complex Component 7/metabolism , N-Glycosyl Hydrolases/metabolism , Protein Binding , Ubiquitin/metabolism , Ubiquitination , Xenopus
2.
Genes Dev ; 31(3): 275-290, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28235849

ABSTRACT

A key event during eukaryotic replication termination is the removal of the CMG helicase from chromatin. CMG unloading involves ubiquitylation of its Mcm7 subunit and the action of the p97 ATPase. Using a proteomic screen in Xenopus egg extracts, we identified factors that are enriched on chromatin when CMG unloading is blocked. This approach identified the E3 ubiquitin ligase CRL2Lrr1, a specific p97 complex, other potential regulators of termination, and many replisome components. We show that Mcm7 ubiquitylation and CRL2Lrr1 binding to chromatin are temporally linked and occur only during replication termination. In the absence of CRL2Lrr1, Mcm7 is not ubiquitylated, CMG unloading is inhibited, and a large subcomplex of the vertebrate replisome that includes DNA Pol ε is retained on DNA. Our data identify CRL2Lrr1 as a master regulator of replisome disassembly during vertebrate DNA replication termination.


Subject(s)
Chromatin/metabolism , DNA Helicases/metabolism , DNA Replication , DNA/metabolism , Ubiquitin-Protein Ligases/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/genetics , Adenosine Triphosphatases/metabolism , Animals , Chromatin/genetics , DNA Polymerase II/metabolism , Minichromosome Maintenance Complex Component 7/metabolism , Nuclear Proteins/metabolism , Ubiquitination , Xenopus Proteins/genetics , Xenopus laevis/metabolism
3.
J Biol Chem ; 298(8): 102234, 2022 08.
Article in English | MEDLINE | ID: mdl-35798141

ABSTRACT

Complex cellular processes are driven by the regulated assembly and disassembly of large multiprotein complexes. While we are beginning to understand the molecular mechanism for assembly of the eukaryotic DNA replication machinery (replisome), we still know relatively little about the regulation of its disassembly at replication termination. Recently, the first elements of this process have emerged, revealing that the replicative helicase, at the heart of the replisome, is polyubiquitylated prior to unloading and that this unloading requires p97 segregase activity. Two different E3 ubiquitin ligases have now been shown to ubiquitylate the helicase under different conditions: Cul2Lrr1 and TRAIP. Here, using Xenopus laevis egg extract cell-free system and biochemical approaches, we have found two p97 cofactors, Ubxn7 and Faf1, which can interact with p97 during replisome disassembly during S-phase. We show only Ubxn7, however, facilitates efficient replisome disassembly. Ubxn7 delivers this role through its interaction via independent domains with both Cul2Lrr1 and p97 to allow coupling between Mcm7 ubiquitylation and its removal from chromatin. Our data therefore characterize Ubxn7 as the first substrate-specific p97 cofactor regulating replisome disassembly in vertebrates and a rationale for the efficacy of the Cul2Lrr1 replisome unloading pathway in unperturbed S-phase.


Subject(s)
Adaptor Proteins, Signal Transducing , Chromatin , DNA Replication , S Phase , Xenopus Proteins , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chromatin/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Minichromosome Maintenance Complex Component 7/metabolism , Ubiquitination , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
4.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: mdl-33834183

ABSTRACT

Minichromosome maintenance complex component 7 (MCM7) belongs to the minichromosome maintenance family that is important for the initiation of eukaryotic DNA replication. Overexpression of the MCM7 protein is relative to cellular proliferation and responsible for aggressive malignancy in various cancers. Mechanistically, inhibition of MCM7 significantly reduces the cellular proliferation associated with cancer. To date, no effective small molecular candidate has been identified that can block the progression of cancer induced by the MCM7 protein. Therefore, the study has been designed to identify small molecular-like natural drug candidates against aggressive malignancy associated with various cancers by targeting MCM7 protein. To identify potential compounds against the targeted protein a comprehensive in silico drug design including molecular docking, ADME (Absorption, Distribution, Metabolism and Excretion), toxicity, and molecular dynamics (MD) simulation approaches has been applied. Seventy phytochemicals isolated from the neem tree (Azadiractha indica) were retrieved and screened against MCM7 protein by using the molecular docking simulation method, where the top four compounds have been chosen for further evaluation based on their binding affinities. Analysis of ADME and toxicity properties reveals the efficacy and safety of the selected four compounds. To validate the stability of the protein-ligand complex structure MD simulations approach has also been performed to the protein-ligand complex structure, which confirmed the stability of the selected three compounds including CAS ID:105377-74-0, CID:12308716 and CID:10505484 to the binding site of the protein. In the study, a comprehensive data screening process has performed based on the docking, ADMET properties, and MD simulation approaches, which found a good value of the selected four compounds against the targeted MCM7 protein and indicates as a promising and effective human anticancer agent.


Subject(s)
Azadirachta/chemistry , Informatics/methods , Minichromosome Maintenance Complex Component 7/antagonists & inhibitors , Molecular Dynamics Simulation , Neoplasms/drug therapy , Phytochemicals/therapeutic use , Algorithms , Binding Sites , Early Detection of Cancer , Humans , Ligands , Minichromosome Maintenance Complex Component 7/chemistry , Minichromosome Maintenance Complex Component 7/metabolism , Molecular Docking Simulation , Molecular Targeted Therapy/methods , Neoplasms/diagnosis , Neoplasms/metabolism , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Plants, Medicinal/chemistry , Protein Binding , Protein Domains , Thermodynamics
5.
EMBO Rep ; 22(3): e52164, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33590678

ABSTRACT

The eukaryotic replisome is disassembled in each cell cycle, dependent upon ubiquitylation of the CMG helicase. Studies of Saccharomyces cerevisiae, Caenorhabditis elegans and Xenopus laevis have revealed surprising evolutionary diversity in the ubiquitin ligases that control CMG ubiquitylation, but regulated disassembly of the mammalian replisome has yet to be explored. Here, we describe a model system for studying the ubiquitylation and chromatin extraction of the mammalian CMG replisome, based on mouse embryonic stem cells. We show that the ubiquitin ligase CUL2LRR1 is required for ubiquitylation of the CMG-MCM7 subunit during S-phase, leading to disassembly by the p97 ATPase. Moreover, a second pathway of CMG disassembly is activated during mitosis, dependent upon the TRAIP ubiquitin ligase that is mutated in primordial dwarfism and mis-regulated in various cancers. These findings indicate that replisome disassembly in diverse metazoa is regulated by a conserved pair of ubiquitin ligases, distinct from those present in other eukaryotes.


Subject(s)
DNA Helicases , DNA Replication , Animals , Cell Cycle/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , Mice , Minichromosome Maintenance Complex Component 7/genetics , Minichromosome Maintenance Complex Component 7/metabolism , Ubiquitination , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
6.
Nucleic Acids Res ; 49(22): 13194-13206, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34850944

ABSTRACT

When vertebrate replisomes from neighboring origins converge, the Mcm7 subunit of the replicative helicase, CMG, is ubiquitylated by the E3 ubiquitin ligase, CRL2Lrr1. Polyubiquitylated CMG is then disassembled by the p97 ATPase, leading to replication termination. To avoid premature replisome disassembly, CRL2Lrr1 is only recruited to CMGs after they converge, but the underlying mechanism is unclear. Here, we use cryogenic electron microscopy to determine structures of recombinant Xenopus laevis CRL2Lrr1 with and without neddylation. The structures reveal that CRL2Lrr1 adopts an unusually open architecture, in which the putative substrate-recognition subunit, Lrr1, is located far from the catalytic module that catalyzes ubiquitin transfer. We further demonstrate that a predicted, flexible pleckstrin homology domain at the N-terminus of Lrr1 is essential to target CRL2Lrr1 to terminated CMGs. We propose a hypothetical model that explains how CRL2Lrr1's catalytic module is positioned next to the ubiquitylation site on Mcm7, and why CRL2Lrr1 binds CMG only after replisomes converge.


Subject(s)
DNA Replication/genetics , Ubiquitin-Protein Ligases/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Animals , Cryoelectron Microscopy , DNA Helicases/genetics , DNA Helicases/metabolism , Minichromosome Maintenance Complex Component 7/genetics , Minichromosome Maintenance Complex Component 7/metabolism , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/ultrastructure , Ubiquitination , Xenopus Proteins/chemistry , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
7.
Curr Genet ; 68(2): 165-179, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35150303

ABSTRACT

Treating yeast cells with the replication inhibitor hydroxyurea activates the S phase checkpoint kinase Rad53, eliciting responses that block DNA replication origin firing, stabilize replication forks, and prevent premature extension of the mitotic spindle. We previously found overproduction of Stn1, a subunit of the telomere-binding Cdc13-Stn1-Ten1 complex, circumvents Rad53 checkpoint functions in hydroxyurea, inducing late origin firing and premature spindle extension even though Rad53 is activated normally. Here, we show Stn1 overproduction acts through remarkably similar pathways compared to loss of RAD53, converging on the MCM complex that initiates origin firing and forms the catalytic core of the replicative DNA helicase. First, mutations affecting Mcm2 and Mcm5 block the ability of Stn1 overproduction to disrupt the S phase checkpoint. Second, loss of function stn1 mutations compensate rad53 S phase checkpoint defects. Third Stn1 overproduction suppresses a mutation in Mcm7. Fourth, stn1 mutants accumulate single-stranded DNA at non-telomeric genome locations, imposing a requirement for post-replication DNA repair. We discuss these interactions in terms of a model in which Stn1 acts as an accessory replication factor that facilitates MCM activation at ORIs and potentially also maintains MCM activity at replication forks advancing through challenging templates.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA Replication/genetics , Minichromosome Maintenance Complex Component 7/genetics , Minichromosome Maintenance Complex Component 7/metabolism , Mutation , Protein Serine-Threonine Kinases , S Phase/genetics , S Phase Cell Cycle Checkpoints/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Telomere-Binding Proteins/metabolism
8.
Mol Cell ; 55(1): 123-37, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24910095

ABSTRACT

NCOA4 is a transcriptional coactivator of nuclear hormone receptors that undergoes gene rearrangement in human cancer. By combining studies in Xenopus laevis egg extracts and mouse embryonic fibroblasts (MEFs), we show here that NCOA4 is a minichromosome maintenance 7 (MCM7)-interacting protein that is able to control DNA replication. Depletion-reconstitution experiments in Xenopus laevis egg extracts indicate that NCOA4 acts as an inhibitor of DNA replication origin activation by regulating CMG (CDC45/MCM2-7/GINS) helicase. NCOA4(-/-) MEFs display unscheduled origin activation and reduced interorigin distance; this results in replication stress, as shown by the presence of fork stalling, reduction of fork speed, and premature senescence. Together, our findings indicate that NCOA4 acts as a regulator of DNA replication origins that helps prevent inappropriate DNA synthesis and replication stress.


Subject(s)
DNA Replication , Nuclear Receptor Coactivators/physiology , Replication Origin , Animals , Cells, Cultured , Cellular Senescence , HeLa Cells , Humans , Mice , Minichromosome Maintenance Complex Component 7/metabolism , Nuclear Receptor Coactivators/metabolism , Two-Hybrid System Techniques , Xenopus laevis
9.
Biochem J ; 478(14): 2825-2842, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34195792

ABSTRACT

Cullin ubiquitin ligases drive replisome disassembly during DNA replication termination. In worm, frog and mouse cells, CUL2LRR1 is required to ubiquitylate the MCM7 subunit of the CMG helicase. Here, we show that cullin ligases also drive CMG-MCM7 ubiquitylation in human cells, thereby making the helicase into a substrate for the p97 unfoldase. Using purified human proteins, including a panel of E2 ubiquitin-conjugating enzymes, we have reconstituted CMG helicase ubiquitylation, dependent upon neddylated CUL2LRR1. The reaction is highly specific to CMG-MCM7 and requires the LRR1 substrate targeting subunit, since replacement of LRR1 with the alternative CUL2 adaptor VHL switches ubiquitylation from CMG-MCM7 to HIF1. CUL2LRR1 firstly drives monoubiquitylation of CMG-MCM7 by the UBE2D class of E2 enzymes. Subsequently, CUL2LRR1 activates UBE2R1/R2 or UBE2G1/G2 to extend a single K48-linked ubiquitin chain on CMG-MCM7. Thereby, CUL2LRR1 converts CMG into a substrate for p97, which disassembles the ubiquitylated helicase during DNA replication termination.


Subject(s)
Cullin Proteins/metabolism , DNA Helicases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Cell Line , Cloning, Molecular/methods , Cullin Proteins/genetics , DNA Helicases/genetics , Humans , Immunoblotting , Lysine/metabolism , Minichromosome Maintenance Complex Component 7/genetics , Minichromosome Maintenance Complex Component 7/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sf9 Cells , Spodoptera , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligases/genetics , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism
10.
Immunopharmacol Immunotoxicol ; 44(1): 17-27, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34821526

ABSTRACT

OBJECTIVE: Aberrant DNA replication is regarded as a component of cancer development. Minichromosome maintenance protein 7 (MCM7), which is critical for the initiation of DNA replication, is overexpressed in multiple malignancies. The effect of MCM7 on cell proliferation, apoptosis, and drug resistance of liver cancer and its mechanism were investigated in this study. METHODS: MCM7 expression in normal liver cells, liver cancer cell lines, and tissues, as well as adjacent tissues, was determined by qRT-PCR. CCK-8 and flow cytometry was performed to detect cell viability, apoptosis, and cell cycle, respectively. The related mRNA and protein expressions were detected by qRT-PCR and western blot. RESULTS: High expression of MCM7 was found in liver cancer tissues and cells, which results in notably lower survival time of patients. Cisplatin (DDP) could inhibit cell proliferation and affect MCM7 expression. Silencing of MCM7 inhibited cell viability, promoted cell apoptosis, arrested cell cycle at G1 phase, and enhanced the effect of DDP on cancer cells, while overexpression of MCM7 did the opposite. Moreover, silencing of MCM7 inhibited cyclinD1 and Ki-67 expressions. The overexpression of MCM7 increased phosphorylation levels of PI3K and AKT, activated the PI3K/AKT pathway, and weakened the inhibitory effect of DDP on the PI3K/AKT pathway. CONCLUSION: Silencing of MCM7 may inhibit cell proliferation and promote apoptosis by regulating the PI3K/AKT pathway to affect the cell cycle, thus affecting the development of liver cancer, and improving the sensitivity of liver cancer cells to DDP.


Subject(s)
Cisplatin , Drug Resistance, Neoplasm/genetics , Liver Neoplasms , Minichromosome Maintenance Complex Component 7 , Signal Transduction , Apoptosis , Cell Line , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Minichromosome Maintenance Complex Component 7/genetics , Minichromosome Maintenance Complex Component 7/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
11.
J Cell Mol Med ; 25(7): 3537-3547, 2021 04.
Article in English | MEDLINE | ID: mdl-33675123

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) is a type of methyltransferase enzyme that can catalyse arginine methylation of histones and non-histone proteins. Accumulating evidence indicates that PRMT5 promotes cancer development and progression. However, its function in colorectal cancer (CRC) is poorly understood. In this study, we revealed the oncogenic roles of PRMT5 in CRC. We found that PRMT5 promoted CRC cell proliferation, migration and invasion in vitro and in vivo. We identified minichromosome maintenance-7 (MCM7) as the direct PRMT5-binding partner. A co-immunoprecipitation (co-IP) assay indicated that PRMT5 physically interacted with MCM7 and that the direct binding domain was located between residues 1-248 in MCM7. In addition, our results from analysis of 99 CRC tissues and 77 adjacent non-cancerous tissues indicated that the PRMT5 and MCM7 expression levels were significantly higher in CRC tissues than in control tissues, which was further confirmed by bioinformatic analysis using TCGA and GEO datasets. We also found that MCM7 promoted CRC cell proliferation, migration and invasion in vitro. Furthermore, we observed that increased PRMT5 expression predicted unfavourable patient survival in CRC patients and in the subgroup of patients with a tumour size of ≤5 cm. These data suggested that PRMT5 and MCM7 might be novel potential targets for the treatment of CRC.


Subject(s)
Carcinogenesis/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Minichromosome Maintenance Complex Component 7/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Animals , Arginine/metabolism , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Knockdown Techniques , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Binding , Protein Interaction Domains and Motifs
12.
Curr Issues Mol Biol ; 43(2): 802-817, 2021 Jul 25.
Article in English | MEDLINE | ID: mdl-34449552

ABSTRACT

Uterine fibroids are the most common mesenchymal uterine neoplasms; their prevalence is estimated in 40%-60% of women under 35 and in 70%-80% of women over 50 years of age. The current research aims to focus on the etiopathogenesis of uterine fibroids, the factors that affect their growth, and markers with diagnostic and prognostic properties. The MCM (minichromosome maintenance) protein family consists of peptides whose primary function is participation in the molecular mechanism of creating replication forks while regulating DNA synthesis. The aim of this work was to determine the proliferative potential of uterine fibroid cells based on the expression of the Ki-67 antigen and the MCMs-i.e., MCM-3, MCM-5, and MCM-7. In addition, the expression of estrogen (ER) and progesterone (PgR) receptors was evaluated and correlated with the expression of the abovementioned observations. Ultimately, received results were analyzed in terms of clinical and pathological data. MATERIALS AND METHODS: In forty-four cases of uterine fibroids, immunohistochemical reactions were performed. A tissue microarray (TMA) technique was utilized and analyzed cases were assessed in triplicate. Immunohistochemistry was performed using antibodies against Ki-67 antigen, ER, PgR, MCM-3, MCM-5, and MCM-8 on an automated staining platform. Reactions were digitalized by a histologic scanner and quantified utilizing dedicated software for nuclear analysis. Assessment was based on quantification expression of the three histiospots, each representing one case in TMA. RESULTS: In the study group (uterine fibroids), statistically significant stronger expression of all the investigated MCMs was observed, as compared to the control group. In addition, moderate and strong positive correlations were found between all tested proliferative markers. The expression of the MCM-7 protein also correlated positively with ER and PgR. With regard to clinical and pathological data, there was a negative correlation between the expression of MCMs and the number of both pregnancies and births. Significant reductions in MCM-5 and MCM-7 expression were observed in the group of women receiving oral hormonal contraceptives, while smoking women showed an increase in MCM-7, ER, and PgR. CONCLUSIONS: Uterine fibroid cells have greater proliferative potential, as evaluated by expression of the Ki-67 antigen and MCMs, than unaltered myometrial cells of the uterine corpus. The expression of MCM-7 was found to have strong or moderate correlations in all assessed relations. In the context of the clinical data, as well evident proliferative potential of MCMs, further studies are strongly recommended.


Subject(s)
Cell Cycle Proteins/biosynthesis , Leiomyoma/metabolism , Minichromosome Maintenance Complex Component 3/biosynthesis , Minichromosome Maintenance Complex Component 7/biosynthesis , Uterine Neoplasms/metabolism , Adult , Aged , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Immunohistochemistry , Leiomyoma/pathology , Middle Aged , Minichromosome Maintenance Complex Component 3/metabolism , Minichromosome Maintenance Complex Component 7/metabolism , Pregnancy , Receptors, Progesterone/metabolism , Uterine Neoplasms/pathology
13.
Mol Cell ; 51(5): 678-90, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23993743

ABSTRACT

Proteins disabled in Fanconi anemia (FA) are necessary for the maintenance of genome stability during cell proliferation. Upon replication stress signaling by ATR, the FA core complex monoubiquitinates FANCD2 and FANCI in order to activate DNA repair. Here, we identified FANCD2 and FANCI in a proteomic screen of replisome-associated factors bound to nascent DNA in response to replication arrest. We found that FANCD2 can interact directly with minichromosome maintenance (MCM) proteins. ATR signaling promoted the transient association of endogenous FANCD2 with the MCM2-MCM7 replicative helicase independently of FANCD2 monoubiquitination. FANCD2 was necessary for human primary cells to restrain DNA synthesis in the presence of a reduced pool of nucleotides and prevented the accumulation of single-stranded DNA, the induction of p21, and the entry of cells into senescence. These data reveal that FANCD2 is an effector of ATR signaling implicated in a general replisome surveillance mechanism that is necessary for sustaining cell proliferation and attenuating carcinogenesis.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/metabolism , Minichromosome Maintenance Complex Component 2/metabolism , Minichromosome Maintenance Complex Component 7/metabolism , S Phase Cell Cycle Checkpoints/physiology , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Proliferation , Cells, Cultured , Cellular Senescence , DNA Replication , Humans , Minichromosome Maintenance Complex Component 2/genetics , Minichromosome Maintenance Complex Component 7/genetics , Signal Transduction/genetics
14.
J Cell Biochem ; 121(2): 1283-1294, 2020 02.
Article in English | MEDLINE | ID: mdl-31535400

ABSTRACT

Cutaneous melanoma (CM) has become a major public health concern. Studies illustrate that minichromosome maintenance protein 7 (MCM7) participate in various diseases including skin disease. Our study aimed to study the effects of MCM7 silencing on CM cell autophagy and apoptosis by modulating the AKT threonine kinase 1 (AKT1)/mechanistic target of rapamycin kinase (mTOR) signaling pathway. Initially, microarray analysis was used to screen the CM-related gene expression data as well as differentially expressed genes. Subsequently, MCM7 expression vector and lentivirus RNA used for MCM7 silencing (LV-shRNA-MCM7) were constructed, and these vectors, dimethyl sulfoxide (DMSO) and AKT activator SC79 were then introduced into CM cell line SK-MEL-2 to validate the role of MCM7 in cell autophagy, viability, apoptosis, cell cycle, migration, and invasion. To further investigate the regulatory mechanisms of MCM7 in CM progress, the expression of MCM7, AKT1, mTOR, cyclin D1, as well as autophagy and apoptosis relative factors, such as LC3B, SOD2, DJ-1, p62, Bcl-2, Bax, and caspase-3 in melanoma cells was determined. MCM7 might mediate the AKT1/mTOR signaling pathway to influence the progress of melanoma. MCM7 silencing contributed to the increased expression of Bax, capase-3, and autophagy-related genes (LC3B, SOD2, and DJ-1), but decreased the expression of Bcl-2, which suggested that MCM7 silencing promoted autophagy and cell apoptosis. At the same time, MCM7 silencing also attenuated cell viability, invasion, and migration, and reduced the cyclin D1 expression and protein levels of p-AKT1 and p-mTOR. Taken together, MCM7 silencing inhibited CM via inactivation of the AKT1/mTOR signaling pathway.


Subject(s)
Autophagy , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/pathology , Minichromosome Maintenance Complex Component 7/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Humans , Melanoma/genetics , Melanoma/metabolism , Minichromosome Maintenance Complex Component 7/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Melanoma, Cutaneous Malignant
15.
Biochem Soc Trans ; 48(3): 823-836, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32490508

ABSTRACT

DNA replication is a complex process that needs to be executed accurately before cell division in order to maintain genome integrity. DNA replication is divided into three main stages: initiation, elongation and termination. One of the key events during initiation is the assembly of the replicative helicase at origins of replication, and this mechanism has been very well described over the last decades. In the last six years however, researchers have also focused on deciphering the molecular mechanisms underlying the disassembly of the replicative helicase during termination. Similar to replisome assembly, the mechanism of replisome disassembly is strictly regulated and well conserved throughout evolution, although its complexity increases in higher eukaryotes. While budding yeast rely on just one pathway for replisome disassembly in S phase, higher eukaryotes evolved an additional mitotic pathway over and above the default S phase specific pathway. Moreover, replisome disassembly has been recently found to be a key event prior to the repair of certain DNA lesions, such as under-replicated DNA in mitosis and inter-strand cross-links (ICLs) in S phase. Although replisome disassembly in human cells has not been characterised yet, they possess all of the factors involved in these pathways in model organisms, and de-regulation of many of them are known to contribute to tumorigenesis and other pathological conditions.


Subject(s)
DNA Replication , Neoplasms/metabolism , S Phase , Saccharomycetales , Animals , Caenorhabditis elegans , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Survival , DNA/metabolism , Genome , Humans , Minichromosome Maintenance Complex Component 7/metabolism , Mitosis , Protein Interaction Mapping , Replication Origin , Saccharomyces cerevisiae , Saccharomycetales/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Xenopus laevis
16.
J Appl Microbiol ; 129(6): 1706-1719, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32320114

ABSTRACT

AIMS: This study evaluates the action of Weissella paramesenteroides WpK4 on amoebic colitis. METHODS AND RESULTS: Weissella paramesenteroides WpK4 was administered in Entamoeba dispar infected and noninfected mice and clinical parameters were evaluated. Following 7 days, the caeca were collected for histopathology, morphometry and immunohistochemical staining of MUC-2, CDC-47 and IgA. The treatment reduced diarrhoea and the presence of blood in the faeces and diminished the area of necrosis, also causing weight gain. Also, the addition of this bacterium enhanced the expression of the mucin (MUC-2). The reduction in necrosis and increased CDC-47 expression indicates significant epithelial regeneration. The negative correlation between CDC-47 and the necrosis area reveals that the bacterium favoured the recovery of the necrotic regions and the positive correlation found between the expression of MUC-2 and CDC-47 indicates that the epithelial regeneration also supports the synthesis of MUC-2. CONCLUSIONS: Weissella paramesenteroides WpK4 was able to increase the protection of the intestinal mucosa against experimental amoebic colitis through the increase of MUC-2 and epithelial regeneration. SIGNIFICANCE AND IMPACT OF THE STUDY: Weissella paramesenteroides WpK4 presents the potential to become a complementary tool in the treatment of amoebic colitis.


Subject(s)
Dysentery, Amebic/prevention & control , Intestinal Mucosa/physiology , Mucin-2/metabolism , Regeneration , Weissella/physiology , Animals , Disease Models, Animal , Dysentery, Amebic/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Mice , Minichromosome Maintenance Complex Component 7/metabolism , Probiotics
17.
Nucleic Acids Res ; 46(13): 6683-6696, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29893900

ABSTRACT

In metazoan cells, only a limited number of mini chromosome maintenance (MCM) complexes are fired during S phase, while the majority remain dormant. Several methods have been used to map replication origins, but such methods cannot identify dormant origins. Herein, we determined MCM7-binding sites in human cells using ChIP-Seq, classified them into firing and dormant origins using origin data and analysed their association with various chromatin signatures. Firing origins, but not dormant origins, were well correlated with open chromatin regions and were enriched upstream of transcription start sites (TSSs) of transcribed genes. Aggregation plots of MCM7 signals revealed minimal difference in the efficacy of MCM loading between firing and dormant origins. We also analysed common fragile sites (CFSs) and found a low density of origins at these sites. Nevertheless, firing origins were enriched upstream of the TSSs. Based on the results, we propose a model in which excessive MCMs are actively loaded in a genome-wide manner, irrespective of chromatin status, but only a fraction are passively fired in chromatin areas with an accessible open structure, such as regions upstream of TSSs of transcribed genes. This plasticity in the specification of replication origins may minimize collisions between replication and transcription.


Subject(s)
Replication Origin , Base Composition , Binding Sites , Chromatin/metabolism , Chromatin Immunoprecipitation , Chromosome Fragile Sites , DNA/chemistry , Genome, Human , HeLa Cells , Humans , Minichromosome Maintenance Complex Component 7/metabolism , Promoter Regions, Genetic , Sequence Analysis, DNA , Transcription Factors/metabolism , Transcription Initiation Site
18.
J Biol Chem ; 292(52): 21417-21430, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29074622

ABSTRACT

In eukaryotes, DNA replication initiates from multiple origins of replication for timely genome duplication. These sites are selected by origin licensing, during which the core enzyme of the eukaryotic DNA replicative helicase, the Mcm2-7 (minichromosome maintenance) complex, is loaded at each origin. This origin licensing requires loading two Mcm2-7 helicases around origin DNA in a head-to-head orientation. Current models suggest that the origin-recognition complex (ORC) and cell-division cycle 6 (Cdc6) proteins recognize and encircle origin DNA and assemble an Mcm2-7 double-hexamer around adjacent double-stranded DNA. To test this model and assess the location of Mcm2-7 initial loading, we placed DNA-protein roadblocks at defined positions adjacent to the essential ORC-binding site within Saccharomyces cerevisiae origin DNA. Roadblocks were made either by covalent cross-linking of the HpaII methyltransferase to DNA or through binding of a transcription activator-like effector (TALE) protein. Contrary to the sites of Mcm2-7 recruitment being precisely defined, only single roadblocks that inhibited ORC-DNA binding showed helicase loading defects. We observed inhibition of helicase loading without inhibition of ORC-DNA binding only when roadblocks were placed on both sides of the origin to restrict sliding of a helicase-loading intermediate. Consistent with a sliding helicase-loading intermediate, when either one of the flanking roadblocks was eliminated, the remaining roadblock had no effect on helicase loading. Interestingly, either origin-flanking nucleosomes or roadblocks resulted in helicase loading being dependent on an additional origin sequence known to be a weaker ORC-DNA-binding site. Together, our findings support a model in which sliding helicase-loading intermediates increase the flexibility of the DNA sequence requirements for origin licensing.


Subject(s)
Arabidopsis Proteins/metabolism , Cell Cycle Proteins/metabolism , Minichromosome Maintenance Proteins/metabolism , Binding Sites , Crystallography, X-Ray , DNA Replication/genetics , DNA Replication/physiology , Minichromosome Maintenance Complex Component 7/metabolism , Minichromosome Maintenance Proteins/physiology , Origin Recognition Complex/genetics , Protein Binding , Protein Domains , Replication Origin/genetics , Replication Origin/physiology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
19.
J Biol Chem ; 292(31): 13008-13021, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28646110

ABSTRACT

The protein mini-chromosome maintenance 10 (Mcm10) was originally identified as an essential yeast protein in the maintenance of mini-chromosome plasmids. Subsequently, Mcm10 has been shown to be required for both initiation and elongation during chromosomal DNA replication. However, it is not fully understood how the multiple functions of Mcm10 are coordinated or how Mcm10 interacts with other factors at replication forks. Here, we identified and characterized the Mcm2-7-interacting domain in human Mcm10. The interaction with Mcm2-7 required the Mcm10 domain that contained amino acids 530-655, which overlapped with the domain required for the stable retention of Mcm10 on chromatin. Expression of truncated Mcm10 in HeLa cells depleted of endogenous Mcm10 via siRNA revealed that the Mcm10 conserved domain (amino acids 200-482) is essential for DNA replication, whereas both the conserved and the Mcm2-7-binding domains were required for its full activity. Mcm10 depletion reduced the initiation frequency of DNA replication and interfered with chromatin loading of replication protein A, DNA polymerase (Pol) α, and proliferating cell nuclear antigen, whereas the chromatin loading of Cdc45 and Pol ϵ was unaffected. These results suggest that human Mcm10 is bound to chromatin through the interaction with Mcm2-7 and is primarily involved in the initiation of DNA replication after loading of Cdc45 and Pol ϵ.


Subject(s)
Chromatin/metabolism , DNA Replication , Minichromosome Maintenance Complex Component 2/metabolism , Minichromosome Maintenance Complex Component 7/metabolism , Minichromosome Maintenance Proteins/metabolism , Origin Recognition Complex/metabolism , Replication Origin , Active Transport, Cell Nucleus , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Minichromosome Maintenance Complex Component 2/chemistry , Minichromosome Maintenance Complex Component 7/chemistry , Minichromosome Maintenance Proteins/antagonists & inhibitors , Minichromosome Maintenance Proteins/chemistry , Minichromosome Maintenance Proteins/genetics , Mutagenesis, Site-Directed , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Multimerization , Protein Stability , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Silent Mutation , Structural Homology, Protein
20.
Biochem Biophys Res Commun ; 506(1): 108-113, 2018 11 17.
Article in English | MEDLINE | ID: mdl-30340827

ABSTRACT

The disassembly of eukaryotic replisome during replication termination is mediated by CRL-dependent poly-ubiquitylation of Mcm7 and p97 segregase. The replisome also disassembles at stalled or collapsed replication forks under certain stress conditions, but the underlying mechanism is poorly understood. Here, we discovered a novel pathway driving stepwise disassembly of the replisome at stalled replication forks after forced entry into M-phase using Xenopus egg extracts. This pathway was dependent on M-CDK activity and K48- and K63-linked poly-ubiquitylation but not on CRL and p97, which is different from known pathways. Furthermore, this pathway could not disassemble converged replisomes whose Mcm7 subunit had been poly-ubiquitylated without p97. These results suggest that there is a distinctive pathway for replisome disassembly when stalled replication forks persist into M-phase.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/genetics , Chromatin/chemistry , DNA Replication , Minichromosome Maintenance Complex Component 7/genetics , Mitosis , Xenopus Proteins/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 3 , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Chromatin/metabolism , Minichromosome Maintenance Complex Component 7/metabolism , Protein Binding , Ubiquitination , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism , Xenopus Proteins/metabolism , Xenopus laevis , Zygote/chemistry , Zygote/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL