Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51.933
Filter
1.
Nat Commun ; 15(1): 5604, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961054

ABSTRACT

The CRL4-DCAF15 E3 ubiquitin ligase complex is targeted by the aryl-sulfonamide molecular glues, leading to neo-substrate recruitment, ubiquitination, and proteasomal degradation. However, the physiological function of DCAF15 remains unknown. Using a domain-focused genetic screening approach, we reveal DCAF15 as an acute myeloid leukemia (AML)-biased dependency. Loss of DCAF15 results in suppression of AML through compromised replication fork integrity and consequent accumulation of DNA damage. Accordingly, DCAF15 loss sensitizes AML to replication stress-inducing therapeutics. Mechanistically, we discover that DCAF15 directly interacts with the SMC1A protein of the cohesin complex and destabilizes the cohesin regulatory factors PDS5A and CDCA5. Loss of PDS5A and CDCA5 removal precludes cohesin acetylation on chromatin, resulting in uncontrolled chromatin loop extrusion, defective DNA replication, and apoptosis. Collectively, our findings uncover an endogenous, cell autonomous function of DCAF15 in sustaining AML proliferation through post-translational control of cohesin dynamics.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Cohesins , DNA Damage , DNA Replication , Leukemia, Myeloid, Acute , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Humans , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Cell Line, Tumor , Acetylation , Animals , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Mice , Chromatin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Apoptosis , Cell Proliferation , HEK293 Cells
2.
Nat Commun ; 15(1): 5727, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977669

ABSTRACT

DNA replication and transcription generate DNA supercoiling, which can cause topological stress and intertwining of daughter chromatin fibers, posing challenges to the completion of DNA replication and chromosome segregation. Type II topoisomerases (Top2s) are enzymes that relieve DNA supercoiling and decatenate braided sister chromatids. How Top2 complexes deal with the topological challenges in different chromatin contexts, and whether all chromosomal contexts are subjected equally to torsional stress and require Top2 activity is unknown. Here we show that catalytic inhibition of the Top2 complex in interphase has a profound effect on the stability of heterochromatin and repetitive DNA elements. Mechanistically, we find that catalytically inactive Top2 is trapped around heterochromatin leading to DNA breaks and unresolved catenates, which necessitate the recruitment of the structure specific endonuclease, Ercc1-XPF, in an SLX4- and SUMO-dependent manner. Our data are consistent with a model in which Top2 complex resolves not only catenates between sister chromatids but also inter-chromosomal catenates between clustered repetitive elements.


Subject(s)
DNA Topoisomerases, Type II , Heterochromatin , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/genetics , Heterochromatin/metabolism , Animals , Topoisomerase II Inhibitors/pharmacology , Repetitive Sequences, Nucleic Acid/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , DNA Replication , DNA, Superhelical/metabolism , DNA, Superhelical/chemistry , Humans , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA/metabolism , DNA/chemistry , Interphase
3.
Nat Commun ; 15(1): 6018, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39019870

ABSTRACT

In Escherichia coli, it is debated whether the two replisomes move independently along the two chromosome arms during replication or if they remain spatially confined. Here, we use high-throughput fluorescence microscopy to simultaneously determine the location and short-time-scale (1 s) movement of the replisome and a chromosomal locus throughout the cell cycle. The assay is performed for several loci. We find that (i) the two replisomes are confined to a region of ~250 nm and ~120 nm along the cell's long and short axis, respectively, (ii) the chromosomal loci move to and through this region sequentially based on their distance from the origin of replication, and (iii) when a locus is being replicated, its short time-scale movement slows down. This behavior is the same at different growth rates. In conclusion, our data supports a model with DNA moving towards spatially confined replisomes at replication.


Subject(s)
Chromosomes, Bacterial , DNA Replication , DNA, Bacterial , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , Chromosomes, Bacterial/genetics , Chromosomes, Bacterial/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Microscopy, Fluorescence , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Replication Origin , Cell Cycle/genetics , DNA-Directed DNA Polymerase , Multienzyme Complexes
4.
Nat Commun ; 15(1): 6039, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39019871

ABSTRACT

During each cell cycle, the process of DNA replication timing is tightly regulated to ensure the accurate duplication of the genome. The extent and significance of alterations in this process during malignant transformation have not been extensively explored. Here, we assess the impact of altered replication timing (ART) on cancer evolution by analysing replication-timing sequencing of cancer and normal cell lines and 952 whole-genome sequenced lung and breast tumours. We find that 6%-18% of the cancer genome exhibits ART, with regions with a change from early to late replication displaying an increased mutation rate and distinct mutational signatures. Whereas regions changing from late to early replication contain genes with increased expression and present a preponderance of APOBEC3-mediated mutation clusters and associated driver mutations. We demonstrate that ART occurs relatively early during cancer evolution and that ART may have a stronger correlation with mutation acquisition than alterations in chromatin structure.


Subject(s)
Breast Neoplasms , DNA Replication Timing , Lung Neoplasms , Mutation , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Cell Line, Tumor , APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism , Mutation Rate , DNA Replication/genetics , Genome, Human
5.
Proc Natl Acad Sci U S A ; 121(28): e2405473121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38950361

ABSTRACT

Cycling cells replicate their DNA during the S phase through a defined temporal program known as replication timing. Mutation frequencies, epigenetic chromatin states, and transcriptional activities are different for genomic regions that are replicated early and late in the S phase. Here, we found from ChIP-Seq analysis that DNA polymerase (Pol) κ is enriched in early-replicating genomic regions in HEK293T cells. In addition, by feeding cells with N 2-heptynyl-2'-deoxyguanosine followed by click chemistry-based enrichment and high-throughput sequencing, we observed elevated Pol κ activities in genomic regions that are replicated early in the S phase. On the basis of the established functions of Pol κ in accurate and efficient nucleotide insertion opposite endogenously induced N 2-modified dG lesions, our work suggests that active engagement of Pol κ may contribute to diminished mutation rates observed in early-replicating regions of the human genome, including cancer genomes. Together, our work expands the functions of Pol κ and offered a plausible mechanism underlying replication timing-dependent mutation accrual in the human genome.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase , S Phase , Humans , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , HEK293 Cells , Genome, Human , DNA Replication Timing
6.
Proc Natl Acad Sci U S A ; 121(28): e2403130121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38950369

ABSTRACT

DNA polymerase κ (Polκ) is a specialized polymerase that has multiple cellular roles such as translesion DNA synthesis, replication of repetitive sequences, and nucleotide excision repair. We have developed a method for capturing DNA synthesized by Polκ utilizing a Polκ-specific substrate, N2-(4-ethynylbenzyl)-2'-deoxyguanosine (EBndG). After shearing of the DNA into 200 to 500 bp lengths, the EBndG-containing DNA was covalently bound to biotin using the Cu(I)-catalyzed alkyne-azide cycloaddition reaction and isolated with streptavidin beads. Isolated DNA was then ligated to adaptors, followed by PCR amplification and next-generation sequencing to generate genome-wide repair maps. We have termed this method polymerase κ sequencing. Here, we present the human genome maps for Polκ activity in an undamaged cell line. We found that Polκ activity was enhanced in GC-rich regions, euchromatin regions, the promoter of genes, and in DNA that is replicated early in the S phase.


Subject(s)
DNA-Directed DNA Polymerase , Fibroblasts , Genome, Human , Humans , DNA-Directed DNA Polymerase/metabolism , Fibroblasts/metabolism , DNA Repair , DNA/metabolism , DNA/genetics , Cell Line , DNA Replication
7.
ACS Chem Biol ; 19(7): 1433-1439, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38959478

ABSTRACT

Most of the human cancers are dependent on telomerase to extend the telomeres. But ∼10% of all cancers use a telomerase-independent, homologous recombination mediated pathway called alternative lengthening of telomeres (ALT). Due to the poor prognosis, ALT status is not being considered yet in the diagnosis of cancer. No such specific treatment is available to date for ALT positive cancers. ALT positive cancers are dependent on replication stress to deploy DNA repair pathways to the telomeres to execute homologous recombination mediated telomere extension. SMARCAL1 (SWI/SNF related, matrix-associated, actin-dependent regulator of chromatin, subfamily A-like 1) is associated with the ALT telomeres to resolve replication stress thus providing telomere stability. Thus, the dependency on replication stress regulatory factors like SMARCAL1 made it a suitable therapeutic target for the treatment of ALT positive cancers. In this study, we found a significant downregulation of SMARCAL1 expression by stabilizing the G-quadruplex (G4) motif found in the promoter of SMARCAL1 by potent G4 stabilizers, like TMPyP4 and BRACO-19. SMARCAL1 downregulation led toward the increased localization of PML (promyelocytic leukemia) bodies in ALT telomeres and triggered the formation of APBs (ALT-associated promyelocytic leukemia bodies) in ALT positive cell lines, increasing telomere replication stress and DNA damage at a genomic level. Induction of replication stress and hyper-recombinogenic phenotype in ALT positive cells mediated by G4 stabilizing molecules already highlighted their possible application as a new therapeutic window to target ALT positive tumors. In accordance with this, our study will also provide a valuable insight toward the development of G4-based ALT therapeutics targeting SMARCAL1.


Subject(s)
DNA Helicases , G-Quadruplexes , Neoplasms , Promoter Regions, Genetic , Telomere , Humans , Telomere/genetics , Telomere/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , Neoplasms/genetics , Cell Line, Tumor , DNA Replication , Telomere Homeostasis
8.
FEMS Microbiol Rev ; 48(4)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38982189

ABSTRACT

Research on Escherichia coli DNA replication paved the groundwork for many breakthrough discoveries with important implications for our understanding of human molecular biology, due to the high level of conservation of key molecular processes involved. To this day, it attracts a lot of attention, partially by virtue of being an important model organism, but also because the understanding of factors influencing replication fidelity might be important for studies on the emergence of antibiotic resistance. Importantly, the wide access to high-resolution single-molecule and live-cell imaging, whole genome sequencing, and cryo-electron microscopy techniques, which were greatly popularized in the last decade, allows us to revisit certain assumptions about the replisomes and offers very detailed insight into how they work. For many parts of the replisome, step-by-step mechanisms have been reconstituted, and some new players identified. This review summarizes the latest developments in the area, focusing on (a) the structure of the replisome and mechanisms of action of its components, (b) organization of replisome transactions and repair, (c) replisome dynamics, and (d) factors influencing the base and sugar fidelity of DNA synthesis.


Subject(s)
DNA Replication , Escherichia coli , Escherichia coli/genetics , Escherichia coli/physiology , Escherichia coli/metabolism , DNA, Bacterial/metabolism , DNA, Bacterial/genetics , DNA Repair
9.
Curr Biol ; 34(14): R680-R682, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39043139

ABSTRACT

Cohesin holds together the sister chromatids from DNA replication onwards. How cohesion is established has long remained a black box. Through recent studies, a model is emerging in which a replisome-cohesin encounter results in the establishment of cohesive linkages at sites of replication termination.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Cohesins , DNA Replication , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatids/metabolism
10.
Nat Commun ; 15(1): 6197, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043663

ABSTRACT

Replication stress compromises genomic integrity. Fork blocking lesions such as those induced by cisplatin and other chemotherapeutic agents arrest replication forks. Repriming downstream of these lesions represents an important mechanism of replication restart, however the single stranded DNA (ssDNA) gaps left behind, unless efficiently filled, can serve as entry point for nucleases. Nascent strand gaps can be repaired by BRCA-mediated homology repair. Alternatively, gaps can also be filled by translesion synthesis (TLS) polymerases. How these events are regulated is still not clear. Here, we show that PARP10, a poorly-characterized mono-ADP-ribosyltransferase, is recruited to nascent strand gaps to promote their repair. PARP10 interacts with the ubiquitin ligase RAD18 and recruits it to these structures, resulting in the ubiquitination of the replication factor PCNA. PCNA ubiquitination, in turn, recruits the TLS polymerase REV1 for gap filling. We show that PARP10 recruitment to gaps and the subsequent REV1-mediated gap filling requires both the catalytic activity of PARP10, and its ability to interact with PCNA. We moreover show that PARP10 is hyperactive in BRCA-deficient cells, and its inactivation potentiates gap accumulations and cytotoxicity in these cells. Our work uncovers PARP10 as a regulator of ssDNA gap filling, which promotes genomic stability in BRCA-deficient cells.


Subject(s)
DNA Repair , DNA Replication , DNA, Single-Stranded , DNA-Binding Proteins , Poly(ADP-ribose) Polymerases , Proliferating Cell Nuclear Antigen , Ubiquitin-Protein Ligases , Ubiquitination , Humans , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/genetics , DNA Damage , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Cell Line, Tumor , HEK293 Cells , Translesion DNA Synthesis , DNA-Directed DNA Polymerase , Proto-Oncogene Proteins
11.
Nat Commun ; 15(1): 6104, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030241

ABSTRACT

G-quadruplexes (G4s) formed by guanine-rich nucleic acids induce genome instability through impeding DNA replication fork progression. G4s are stable DNA structures, the unfolding of which require the functions of DNA helicases. Pif1 helicase binds preferentially to G4 DNA and plays multiple roles in maintaining genome stability, but the mechanism by which Pif1 unfolds G4s is poorly understood. Here we report the co-crystal structure of Saccharomyces cerevisiae Pif1 (ScPif1) bound to a G4 DNA with a 5' single-stranded DNA (ssDNA) segment. Unlike the Thermus oshimai Pif1-G4 structure, in which the 1B and 2B domains confer G4 recognition, ScPif1 recognizes G4 mainly through the wedge region in the 1A domain that contacts the 5' most G-tetrad directly. A conserved Arg residue in the wedge is required for Okazaki fragment processing but not for mitochondrial function or for suppression of gross chromosomal rearrangements. Multiple substitutions at this position have similar effects on resolution of DNA duplexes and G4s, suggesting that ScPif1 may use the same wedge to unwind G4 and dsDNA. Our results reveal the mechanism governing dsDNA unwinding and G4 unfolding by ScPif1 helicase that can potentially be generalized to other eukaryotic Pif1 helicases and beyond.


Subject(s)
DNA Helicases , G-Quadruplexes , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , DNA Helicases/metabolism , DNA Helicases/chemistry , DNA Helicases/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , DNA/metabolism , DNA/chemistry , DNA/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Binding , DNA Replication , Genomic Instability
12.
PLoS Genet ; 20(7): e1011341, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38954736

ABSTRACT

The drug floxuridine (5-fluorodeoxyuridine, FUdR) is an active metabolite of 5-Fluorouracil (5-FU). It converts to 5-fluorodeoxyuridine monophosphate (FdUMP) and 5-fluorodeoxyuridine triphosphate (FdUTP), which on incorporation into the genome inhibits DNA replication. Additionally, it inhibits thymidylate synthase, causing dTMP shortage while increasing dUMP availability, which induces uracil incorporation into the genome. However, the mechanisms underlying cellular tolerance to FUdR are yet to be fully elucidated. In this study, we explored the mechanisms underlying cellular resistance to FUdR by screening for FUdR hypersensitive mutants from a collection of DT40 mutants deficient in each genomic maintenance system. We identified REV3, which is involved in translesion DNA synthesis (TLS), to be a critical factor in FUdR tolerance. Replication using a FUdR-damaged template was attenuated in REV3-/- cells, indicating that the TLS function of REV3 is required to maintain replication on the FUdR-damaged template. Notably, FUdR-exposed REV3-/- cells exhibited defective cell cycle arrest in the early S phase, suggesting that REV3 is involved in intra-S checkpoint activation. Furthermore, REV3-/- cells showed defects in Chk1 phosphorylation, which is required for checkpoint activation, but the survival of FUdR-exposed REV3-/- cells was further reduced by the inhibition of Chk1 or ATR. These data indicate that REV3 mediates DNA checkpoint activation at least through Chk1 phosphorylation, but this signal acts in parallel with ATR-Chk1 DNA damage checkpoint pathway. Collectively, we reveal a previously unappreciated role of REV3 in FUdR tolerance.


Subject(s)
DNA Damage , DNA Replication , Floxuridine , Floxuridine/pharmacology , Animals , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 1/genetics , S Phase Cell Cycle Checkpoints/genetics , S Phase Cell Cycle Checkpoints/drug effects , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Chickens , Humans , DNA Repair/genetics , Phosphorylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Translesion DNA Synthesis , Deoxyuridine/analogs & derivatives
13.
Methods Mol Biol ; 2819: 625-653, 2024.
Article in English | MEDLINE | ID: mdl-39028527

ABSTRACT

Computational models of cells cannot be considered complete unless they include the most fundamental process of life, the replication of genetic material. In a recent study, we presented a computational framework to model systems of replicating bacterial chromosomes as polymers at 10 bp resolution with Brownian dynamics. This approach was used to investigate changes in chromosome organization during replication and extend the applicability of an existing whole-cell model (WCM) for a genetically minimal bacterium, JCVI-syn3A, to the entire cell cycle. To achieve cell-scale chromosome structures that are realistic, we modeled the chromosome as a self-avoiding homopolymer with bending and torsional stiffnesses that capture the essential mechanical properties of dsDNA in Syn3A. Additionally, the polymer interacts with ribosomes distributed according to cryo-electron tomograms of Syn3A. The polymer model was further augmented by computational models of loop extrusion by structural maintenance of chromosomes (SMC) protein complexes and topoisomerase action, and the modeling and analysis of multi-fork replication states.


Subject(s)
Chromosomes, Bacterial , DNA Replication , Chromosomes, Bacterial/genetics , DNA, Bacterial/genetics , Bacteria/genetics
14.
Cell Syst ; 15(7): 639-648.e2, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38981487

ABSTRACT

Systems like the prototypical lac operon can reliably hold repression of transcription upon DNA replication across cell cycles with just 10 repressor molecules per cell and behave as if they were at equilibrium. The origin of this phenomenology is still an unresolved question. Here, we develop a general theory to analyze strong perturbations in quasi-equilibrium systems and use it to quantify the effects of DNA replication in gene regulation. We find a scaling law linking actual with predicted equilibrium transcription via a single kinetic parameter. We show that even the lac operon functions beyond the physical limits of naive regulation through compensatory mechanisms that suppress non-equilibrium effects. Synthetic systems without adjuvant activators, such as the cAMP receptor protein (CRP), lack this reliability. Our results provide a rationale for the function of CRP, beyond just being a tunable activator, as a mitigator of cell cycle perturbations.


Subject(s)
Cell Cycle , DNA Replication , Cell Cycle/genetics , Lac Operon , Gene Expression Regulation , Cyclic AMP Receptor Protein/metabolism , Cyclic AMP Receptor Protein/genetics , Transcription, Genetic , Escherichia coli/genetics , Escherichia coli/metabolism , Kinetics
15.
Genes Dev ; 38(11-12): 504-527, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38986581

ABSTRACT

Genome integrity relies on the accuracy of DNA metabolism, but as appreciated for more than four decades, transcription enhances mutation and recombination frequencies. More recent research provided evidence for a previously unforeseen link between RNA and DNA metabolism, which is often related to the accumulation of DNA-RNA hybrids and R-loops. In addition to physiological roles, R-loops interfere with DNA replication and repair, providing a molecular scenario for the origin of genome instability. Here, we review current knowledge on the multiple RNA factors that prevent or resolve R-loops and consequent transcription-replication conflicts and thus act as modulators of genome dynamics.


Subject(s)
Genomic Instability , R-Loop Structures , RNA , Genomic Instability/genetics , RNA/metabolism , RNA/genetics , DNA Replication/genetics , Animals , Humans , Transcription, Genetic/genetics
16.
Biochem J ; 481(14): 923-944, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38985307

ABSTRACT

Maintenance of genome stability is of paramount importance for the survival of an organism. However, genomic integrity is constantly being challenged by various endogenous and exogenous processes that damage DNA. Therefore, cells are heavily reliant on DNA repair pathways that have evolved to deal with every type of genotoxic insult that threatens to compromise genome stability. Notably, inherited mutations in genes encoding proteins involved in these protective pathways trigger the onset of disease that is driven by chromosome instability e.g. neurodevelopmental abnormalities, neurodegeneration, premature ageing, immunodeficiency and cancer development. The ability of cells to regulate the recruitment of specific DNA repair proteins to sites of DNA damage is extremely complex but is primarily mediated by protein post-translational modifications (PTMs). Ubiquitylation is one such PTM, which controls genome stability by regulating protein localisation, protein turnover, protein-protein interactions and intra-cellular signalling. Over the past two decades, numerous ubiquitin (Ub) E3 ligases have been identified to play a crucial role not only in the initiation of DNA replication and DNA damage repair but also in the efficient termination of these processes. In this review, we discuss our current understanding of how different Ub E3 ligases (RNF168, TRAIP, HUWE1, TRIP12, FANCL, BRCA1, RFWD3) function to regulate DNA repair and replication and the pathological consequences arising from inheriting deleterious mutations that compromise the Ub-dependent DNA damage response.


Subject(s)
DNA Damage , DNA Repair , DNA Replication , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Neoplasms/genetics , Neoplasms/metabolism , Genomic Instability , Protein Processing, Post-Translational , Animals , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
Arch Insect Biochem Physiol ; 116(3): e22132, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38993002

ABSTRACT

Perilipins are evolutionarily conserved from insects to mammals. Drosophila lipid storage droplet-1 (LSD-1) is a lipid storage droplet membrane surface-binding protein family member and a counterpart to mammalian perilipin 1 and is known to play a role in lipolysis. However, the function of LSD-1 during specific tissue development remains under investigation. This study demonstrated the role of LSD-1 in salivary gland development. Knockdown of Lsd-1 in the salivary gland was established using the GAL4/UAS system. The third-instar larvae of knockdown flies had small salivary glands containing cells with smaller nuclei. The null mutant Drosophila also showed the same phenotype. The depletion of LSD-1 expression induced a delay of endoreplication due to decreasing CycE expression and increasing DNA damage. Lsd-1 genetically interacted with Myc in the third-instar larvae. These results demonstrate that LSD-1 is involved in cell cycle and cell death programs in the salivary gland, providing novel insight into the effects of LSD-1 in regulating salivary gland development and the interaction between LSD-1 and Myc.


Subject(s)
Cell Death , Drosophila Proteins , Larva , Salivary Glands , Animals , Salivary Glands/metabolism , Salivary Glands/cytology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Larva/growth & development , Larva/metabolism , Larva/genetics , Drosophila/metabolism , Drosophila/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/growth & development , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , DNA Replication , DNA-Binding Proteins , Oxidoreductases, N-Demethylating , Transcription Factors
18.
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
19.
Postepy Biochem ; 70(1): 8-21, 2024 05 23.
Article in English | MEDLINE | ID: mdl-39016227

ABSTRACT

Genome replication requires duplication of the complete set of DNA sequences together with nucleosomes and epigenetic signatures. Notwithstanding profound knowledge on mechanistic details of DNA replication, major problems of genome replication have remained unresolved. In this perspective article, we consider the accessibility of replication machines to all DNA sequences in due course, the maintenance of functionally important positional and structural features of chromatid domains during replication, and the rapid transition of CTs into prophase chromosomes with two chromatids. We illustrate this problem with EdU pulse-labeling (10 min) and chase experiments (80 min) performed with mouse myeloblast cells. Following light optical serial sectioning of nuclei with 3D structured illumination microscopy (SIM), seven DNA intensity classes were distinguished as proxies for increasing DNA compaction. In nuclei of cells fixed immediately after the pulse-label, we observed a relative under-representation of EdU-labeled DNA in low DNA density classes, representing the active nuclear compartment (ANC), and an over-representation in high density classes representing the inactive nuclear compartment (INC). Cells fixed after the chase revealed an even more pronounced shift to high DNA intensity classes. This finding contrasts with previous studies of the transcriptional topography demonstrating an under-representation of epigenetic signatures for active chromatin and RNAPII in high DNA intensity classes and their over-representation in low density classes. We discuss these findings in the light of current models viewing CDs either as structural chromatin frameworks or as phase-separated droplets, as well as methodological limitations that currently prevent an integration of this contrasting evidence for the spatial nuclear topography of replication and transcription into a common framework of the dynamic nuclear architecture.


Subject(s)
DNA Replication , Animals , Mice , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA/genetics , DNA Replication/physiology , Epigenesis, Genetic/physiology , Genome/genetics , Microscopy/methods
20.
Sci Adv ; 10(29): eado5398, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39018394

ABSTRACT

Initiation of DNA replication in Escherichia coli is coupled to cell size via the DnaA protein, whose activity is dependent on its nucleotide-bound state. However, the oscillations in DnaA activity have never been observed at the single-cell level. By measuring the volume-specific production rate of a reporter protein under control of a DnaA-regulated promoter, we could distinguish two distinct cell-cycle oscillators. The first, driven by both DnaA activity and SeqA repression, shows a causal relationship with cell size and divisions, similarly to initiation events. The second one, a reporter of DnaA activity alone, loses the synchrony and causality properties. Our results show that transient inhibition of gene expression by SeqA keeps the oscillation of volume-sensing DnaA activity in phase with the subsequent division event and suggest that DnaA activity peaks do not correspond directly to initiation events.


Subject(s)
Cell Cycle , Escherichia coli Proteins , Escherichia coli , Gene Expression Regulation, Bacterial , Single-Cell Analysis , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Cell Cycle/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , DNA Replication , Promoter Regions, Genetic , Bacterial Outer Membrane Proteins
SELECTION OF CITATIONS
SEARCH DETAIL