Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.792
Filter
1.
bioRxiv ; 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39282338

ABSTRACT

The CMG helicase (CDC45-MCM2-7-GINS) unwinds DNA as a component of eukaryotic replisomes. Replisome (dis)assembly is tightly coordinated with cell cycle progression to ensure genome stability. However, factors that prevent premature CMG unloading and replisome disassembly are poorly described. Since disassembly is catalyzed by ubiquitination, deubiquitinases (DUBs) represent attractive candidates for safeguarding against untimely and deleterious CMG unloading. We combined a targeted loss-of-function screen with quantitative, single-cell analysis to identify human USP37 as a key DUB preventing replisome disassembly. We demonstrate that USP37 maintains active replisomes on S-phase chromatin and promotes normal cell cycle progression. Proteomics and enzyme assays revealed USP37 interacts with the CMG complex to deubiquitinate MCM7, thus antagonizing replisome disassembly. Significantly, USP37 protects normal epithelial cells from oncoprotein-induced replication stress. Our findings reveal USP37 to be critical to the maintenance of replisomes in S-phase and suggest USP37-targeting as a potential strategy for treating malignancies with defective DNA replication control.

2.
Int J Biol Sci ; 20(12): 4978-4998, 2024.
Article in English | MEDLINE | ID: mdl-39309429

ABSTRACT

Gastric cancer (GC) ranks as the fifth most common cancer and the fourth leading cause of cancer-related deaths globally. Despite advancements in molecular profiling, the mechanisms driving GC proliferation and metastasis remain unclear. This study identifies Early 2 Factor 4 (E2F4) as a key transcription factor that promotes GC cell proliferation, migration, and invasion by upregulating DNA Replication and Sister Chromatid Cohesion 1 (DSCC1) expression. Bioinformatics and transcription factor analyses revealed E2F4 as a significant regulator of DSCC1. Functional assays confirmed E2F4's role in enhancing GC cell malignancy in vitro and in vivo. Knockdown and overexpression experiments demonstrated that E2F4 positively regulates DSCC1 at the transcriptional level, with ChIP-qPCR and dual luciferase reporter assays validating the binding sites on the DSCC1 promoter. These findings highlight the E2F4-DSCC1 axis as a potential therapeutic target to mitigate GC progression.


Subject(s)
Cell Movement , Cell Proliferation , E2F4 Transcription Factor , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Humans , Cell Proliferation/genetics , Cell Movement/genetics , Cell Line, Tumor , E2F4 Transcription Factor/metabolism , E2F4 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Animals , Mice , Mice, Nude , Neoplasm Invasiveness
3.
Plant Cell Rep ; 43(10): 232, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39283352

ABSTRACT

KEY MESSAGE: We used marker-free technologies to study chromatin at cellular resolution. Our results show asymmetric chromatin distribution, explore chromatin dynamics during mitosis, and reveal structural differences between trichoblast and atrichoblast cell. The shapes, sizes, and structural organizations of plant nuclei vary considerably among cell types, tissues, and species. This diversity is dependent on various factors, including cellular function, developmental stage, and environmental or physiological conditions. The differences in nuclear structure reflect the state of chromatin, which, in turn, controls gene expression and regulates cell fate. To examine the interrelationship between nuclear structure, cell morphology, and tissue-specific cell proliferation and differentiation processes, we conducted multiple visualizations of H3K4me1, H3K9me2, 4',6-diamidino-2-phenylindole, 5-ethynyl 2'-deoxyuridine, and SCRI Renaissance 2200, followed by subsequent quantitative analysis of individual cells and nuclei. By assigning cylindrical coordinates to the nuclei in the iRoCS toolbox, we were able to construct in situ digital three-dimensional chromatin maps for all the tissue layers of individual roots. A detailed analysis of the nuclei features of H3K4me1 and H3K9me2 in the mitotic and the elongation zones in trichoblast and atrichoblast cells at the root apical meristem revealed cell type-specific chromatin dynamics with asymmetric distribution of euchromatin and heterochromatin marks that may be associated with cell cycle and cell differentiation characteristics of specific cells. Furthermore, the spatial distribution of nuclei stained with 5-ethynyl 2'-deoxyuridine in the epidermis and cortex tissues suggests short-range coordination of cell division and nuclear migration in a linear sequence through an unknown regulatory mechanism.


Subject(s)
Arabidopsis , Cell Differentiation , Cell Division , Cell Nucleus , Chromatin , Meristem , Meristem/cytology , Meristem/genetics , Chromatin/metabolism , Arabidopsis/cytology , Arabidopsis/genetics , Cell Nucleus/metabolism , Histones/metabolism , Mitosis , Plant Roots/cytology , Plant Roots/genetics
4.
Cell ; 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39293447

ABSTRACT

The genome duplication program is affected by multiple factors in vivo, including developmental cues, genotoxic stress, and aging. Here, we monitored DNA replication initiation dynamics in regenerating livers of young and old mice after partial hepatectomy to investigate the impact of aging. In young mice, the origin firing sites were well defined; the majority were located 10-50 kb upstream or downstream of expressed genes, and their position on the genome was conserved in human cells. Old mice displayed the same replication initiation sites, but origin firing was inefficient and accompanied by a replication stress response. Inhibitors of the ATR checkpoint kinase fully restored origin firing efficiency in the old mice but at the expense of an inflammatory response and without significantly enhancing the fraction of hepatocytes entering the cell cycle. These findings unveil aging-dependent replication stress and a crucial role of ATR in mitigating the stress-associated inflammation, a hallmark of aging.

5.
Cell Rep ; 43(9): 114700, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39235941

ABSTRACT

Genomic imprinting involves differential DNA methylation and gene expression between homologous paternal and maternal loci. It remains unclear, however, whether DNA replication also shows parent-of-origin-specific patterns at imprinted or other genomic regions. Here, we investigate genome-wide asynchronous DNA replication utilizing uniparental human embryonic stem cells containing either maternal-only (parthenogenetic) or paternal-only (androgenetic) DNA. Four clusters of imprinted genes exhibited differential replication timing based on parent of origin, while the remainder of the genome, 99.82%, showed no significant replication asynchrony between parental origins. Active alleles in imprinted gene clusters replicated earlier than their inactive counterparts. At the Prader-Willi syndrome locus, replication asynchrony spanned virtually the entirety of S phase. Replication asynchrony was carried through differentiation to neuronal precursor cells in a manner consistent with gene expression. This study establishes asynchronous DNA replication as a hallmark of large imprinted gene clusters.

6.
Biomol NMR Assign ; 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39269602

ABSTRACT

The 81 kDa E. coli ß clamp is a ring-shaped head-to-tail homodimer that encircles DNA and plays a central role in bacterial DNA replication by serving as a processivity factor for DNA polymerases and a binding platform for other DNA replication and repair proteins. Here we report the backbone 1H, 15N, and 13C NMR resonance assignments of the stabilized T45R/S107R ß clamp variant obtained using standard TROSY-based triple-resonance experiments (BMRB 52548). The backbone assignments were aided by 13C and 15N edited NOESY experiments, allowing us to utilize our previously reported assignments of the ß clamp ILV side-chain methyl groups (BMRB 51430, 51431). The backbone assignments of the T45R/S107R ß clamp variant were transferred to the wild-type ß clamp using a minimal set of TROSY-based 15N edited NOESY, NHCO and NHCA experiments (BMRB 52549). The reported backbone and previous ILV side-chain resonance assignments will enable NMR studies of the ß clamp interactions and dynamics using amide and methyl groups as probes.

7.
DNA Repair (Amst) ; 142: 103758, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39236419

ABSTRACT

Timely and accurate DNA replication is critical for safeguarding genome integrity and ensuring cell viability. Yet, this process is challenged by DNA damage blocking the progression of the replication machinery. To counteract replication fork stalling, evolutionary conserved DNA damage tolerance (DDT) mechanisms promote DNA damage bypass and fork movement. One of these mechanisms involves "skipping" DNA damage through repriming downstream of the lesion, leaving single-stranded DNA (ssDNA) gaps behind the advancing forks (also known as post-replicative gaps). In vertebrates, repriming in damaged leading templates is proposed to be mainly promoted by the primase and polymerase PRIMPOL. In this review, we discuss recent advances towards our understanding of the physiological and pathological conditions leading to repriming activation in human models, revealing a regulatory network of PRIMPOL activity. Upon repriming by PRIMPOL, post-replicative gaps formed can be filled-in by the DDT mechanisms translesion synthesis and template switching. We discuss novel findings on how these mechanisms are regulated and coordinated in time to promote gap filling. Finally, we discuss how defective gap filling and aberrant gap expansion by nucleases underlie the cytotoxicity associated with post-replicative gap accumulation. Our increasing knowledge of this repriming mechanism - from gap formation to gap filling - is revealing that targeting the last step of this pathway is a promising approach to exploit post-replicative gaps in anti-cancer therapeutic strategies.


Subject(s)
DNA Damage , DNA Primase , DNA Replication , DNA-Directed DNA Polymerase , Humans , DNA Primase/metabolism , DNA-Directed DNA Polymerase/metabolism , Animals , DNA Repair , Multifunctional Enzymes/metabolism , DNA, Single-Stranded/metabolism
8.
Cell ; 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39265577

ABSTRACT

DNA repair and autophagy are distinct biological processes vital for cell survival. Although autophagy helps maintain genome stability, there is no evidence of its direct role in the repair of DNA lesions. We discovered that lysosomes process topoisomerase 1 cleavage complexes (TOP1cc) DNA lesions in vertebrates. Selective degradation of TOP1cc by autophagy directs DNA damage repair and cell survival at clinically relevant doses of topoisomerase 1 inhibitors. TOP1cc are exported from the nucleus to lysosomes through a transient alteration of the nuclear envelope and independent of the proteasome. Mechanistically, the autophagy receptor TEX264 acts as a TOP1cc sensor at DNA replication forks, triggering TOP1cc processing by the p97 ATPase and mediating the delivery of TOP1cc to lysosomes in an MRE11-nuclease- and ATR-kinase-dependent manner. We found an evolutionarily conserved role for selective autophagy in DNA repair that enables cell survival, protects genome stability, and is clinically relevant for colorectal cancer patients.

9.
Proteins ; 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39230358

ABSTRACT

In bacteria, chromosome replication is achieved by the coordinations of more than a dozen replisome enzymes. Replication initiation protein DnaA melts DNA duplex at replication origin (oriC) and forms a replication bubble, followed by loading of helicase DnaB with the help of loader protein DnaC. Then the DnaB helicase unwinds the dsDNA and supports the priming of DnaG and the polymerizing of DNA polymerase. The DnaB helicase functions as a platform coupling unwinding, priming, and polymerizing events. The multiple roles of DnaB helicase are underlined by its distinctive architecture and dynamics conformations. In this review, we will discuss the assembling of DnaB hexamer and the conformational changes upon binding of various partners, DnaB in states of closed dilated (CD), closed constricted (CC), closed helical (CH), and open helical (OH) are discussed. These multiple interfaces among DnaB and partners are potential targets for inhibitors design and novel peptide antibiotics development.

10.
DNA Repair (Amst) ; 142: 103741, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39153403

ABSTRACT

PrimPol is a human DNA primase involved in DNA damage tolerance pathways by restarting DNA replication downstream of DNA lesions and non-canonical DNA structures. Activity and affinity to DNA relays on the interaction of PrimPol with replication protein A (RPA). In this work, we report that PrimPol has an intrinsic ability to copy DNA hairpins with a stem length of 5-9 base pairs (bp) but shows pronounced pausing of DNA synthesis. RPA greatly stimulates DNA synthesis across inverted DNA repeats by PrimPol. Moreover, deletion of the C-terminal RPA binding motif (RBM) facilitates DNA hairpin bypass and makes it independent of RPA. This work supports the idea that RBM is a negative regulator of PrimPol and its interaction with RPA is required to achieve the fully active state.


Subject(s)
DNA Primase , DNA Replication , DNA , Humans , DNA Primase/metabolism , DNA Primase/chemistry , DNA Primase/genetics , DNA/metabolism , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/chemistry , Replication Protein A/metabolism , Nucleic Acid Conformation , DNA-Directed DNA Polymerase/metabolism , Inverted Repeat Sequences , Protein Binding
11.
J Biol Chem ; 300(9): 107688, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39159820

ABSTRACT

Ribonucleotides in DNA cause several types of genome instability and can be removed by ribonucleotide excision repair (RER) that is finalized by DNA ligase 1 (LIG1). However, the mechanism by which LIG1 discriminates the RER intermediate containing a 5'-RNA-DNA lesion generated by RNase H2-mediated cleavage of ribonucleotides at atomic resolution remains unknown. Here, we determine X-ray structures of LIG1/5'-rG:C at the initial step of ligation where AMP is bound to the active site of the ligase and uncover a large conformational change downstream the nick resulting in a shift at Arg(R)871 residue in the Adenylation domain of the ligase. Furthermore, we demonstrate a diminished ligation of the nick DNA substrate with a 5'-ribonucleotide in comparison to an efficient end joining of the nick substrate with a 3'-ribonucleotide by LIG1. Finally, our results demonstrate that mutations at the active site residues of the ligase and LIG1 disease-associated variants significantly impact the ligation efficiency of RNA-DNA heteroduplexes harboring "wrong" sugar at 3'- or 5'-end of nick. Collectively, our findings provide a novel atomic insight into proficient sugar discrimination by LIG1 during the processing of the most abundant form of DNA damage in cells, genomic ribonucleotides, during the initial step of the RER pathway.

12.
Mol Cell ; 84(16): 3044-3060.e11, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39142279

ABSTRACT

G-quadruplexes (G4s) form throughout the genome and influence important cellular processes. Their deregulation can challenge DNA replication fork progression and threaten genome stability. Here, we demonstrate an unexpected role for the double-stranded DNA (dsDNA) translocase helicase-like transcription factor (HLTF) in responding to G4s. We show that HLTF, which is enriched at G4s in the human genome, can directly unfold G4s in vitro and uses this ATP-dependent translocase function to suppress G4 accumulation throughout the cell cycle. Additionally, MSH2 (a component of MutS heterodimers that bind G4s) and HLTF act synergistically to suppress G4 accumulation, restrict alternative lengthening of telomeres, and promote resistance to G4-stabilizing drugs. In a discrete but complementary role, HLTF restrains DNA synthesis when G4s are stabilized by suppressing primase-polymerase (PrimPol)-dependent repriming. Together, the distinct roles of HLTF in the G4 response prevent DNA damage and potentially mutagenic replication to safeguard genome stability.


Subject(s)
DNA Primase , DNA Replication , DNA-Binding Proteins , G-Quadruplexes , Genomic Instability , MutS Homolog 2 Protein , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , MutS Homolog 2 Protein/metabolism , MutS Homolog 2 Protein/genetics , DNA Primase/metabolism , DNA Primase/genetics , Telomere Homeostasis , DNA Damage , HEK293 Cells , Multifunctional Enzymes/metabolism , Multifunctional Enzymes/genetics , DNA-Directed DNA Polymerase
13.
Proc Natl Acad Sci U S A ; 121(34): e2322938121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39141351

ABSTRACT

The removal of mis-incorporated nucleotides by proofreading activity ensures DNA replication fidelity. Whereas the ε-exonuclease DnaQ is a well-established proofreader in the model organism Escherichia coli, it has been shown that proofreading in a majority of bacteria relies on the polymerase and histidinol phosphatase (PHP) domain of replicative polymerase, despite the presence of a DnaQ homolog that is structurally and functionally distinct from E. coli DnaQ. However, the biological functions of this type of noncanonical DnaQ remain unclear. Here, we provide independent evidence that noncanonical DnaQ functions as an additional proofreader for mycobacteria. Using the mutation accumulation assay in combination with whole-genome sequencing, we showed that depletion of DnaQ in Mycolicibacterium smegmatis leads to an increased mutation rate, resulting in AT-biased mutagenesis and increased insertions/deletions in the homopolymer tract. Our results showed that mycobacterial DnaQ binds to the ß clamp and functions synergistically with the PHP domain proofreader to correct replication errors. Furthermore, the loss of dnaQ results in replication fork dysfunction, leading to attenuated growth and increased mutagenesis on subinhibitory fluoroquinolones potentially due to increased vulnerability to fork collapse. By analyzing the sequence polymorphism of dnaQ in clinical isolates of Mycobacterium tuberculosis (Mtb), we demonstrated that a naturally evolved DnaQ variant prevalent in Mtb lineage 4.3 may enable hypermutability and is associated with drug resistance. These results establish a coproofreading model and suggest a division of labor between DnaQ and PHP domain proofreader. This study also provides real-world evidence that a mutator-driven evolutionary pathway may exist during the adaptation of Mtb.


Subject(s)
DNA Replication , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Mutation
14.
Cell ; 187(18): 5029-5047.e21, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39094569

ABSTRACT

The inheritance of parental histones across the replication fork is thought to mediate epigenetic memory. Here, we reveal that fission yeast Mrc1 (CLASPIN in humans) binds H3-H4 tetramers and operates as a central coordinator of symmetric parental histone inheritance. Mrc1 mutants in a key connector domain disrupted segregation of parental histones to the lagging strand comparable to Mcm2 histone-binding mutants. Both mutants showed clonal and asymmetric loss of H3K9me-mediated gene silencing. AlphaFold predicted co-chaperoning of H3-H4 tetramers by Mrc1 and Mcm2, with the Mrc1 connector domain bridging histone and Mcm2 binding. Biochemical and functional analysis validated this model and revealed a duality in Mrc1 function: disabling histone binding in the connector domain disrupted lagging-strand recycling while another histone-binding mutation impaired leading strand recycling. We propose that Mrc1 toggles histones between the lagging and leading strand recycling pathways, in part by intra-replisome co-chaperoning, to ensure epigenetic transmission to both daughter cells.


Subject(s)
DNA Replication , Epigenesis, Genetic , Histones , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Histones/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Mutation , Epigenetic Memory
15.
Cell Rep ; 43(8): 114594, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39116203

ABSTRACT

Homologous recombination (HR) plays an essential role in the repair of DNA double-strand breaks (DSBs), replication stress responses, and genome maintenance. However, unregulated HR during replication can impair genome duplication and compromise genome stability. The mechanisms underlying HR regulation during DNA replication are obscure. Here, we find that RTEL1 helicase, RAD51, and RAD51 paralogs are enriched at stalled replication sites. The absence of RTEL1 leads to an increase in the RAD51-mediated HR and fork reversal during replication and affects genome-wide replication, which can be rescued by co-depleting RAD51 and RAD51 paralogs. Interestingly, co-depletion of fork remodelers such as SMARCAL1/ZRANB3/HLTF/FBH1 and expression of HR-defective RAD51 mutants also rescues replication defects in RTEL1-deficient cells. The anti-recombinase function of RTEL1 during replication depends on its interaction with PCNA and helicase activity. Together, our data identify the role of RTEL1 helicase in restricting RAD51-mediated fork reversal and HR activity to facilitate error-free genome duplication.


Subject(s)
DNA Helicases , DNA Replication , Homologous Recombination , Rad51 Recombinase , Rad51 Recombinase/metabolism , Rad51 Recombinase/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Humans , Proliferating Cell Nuclear Antigen/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA Breaks, Double-Stranded , Genomic Instability
16.
J Mol Biol ; 436(20): 168744, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39147125

ABSTRACT

DNA modified with C2'-methoxy (C2'-OMe) greatly enhances its resistance to nucleases, which is beneficial for the half-life of aptamers and DNA nanomaterials. Although the unnatural DNA polymerases capable of incorporating C2'-OMe modified nucleoside monophosphates (C2'-OMe-NMPs) were engineered via directed evolution, the detailed molecular mechanism by which an evolved DNA polymerase recognizes C2'-OMe-NTPs remains poorly understood. Here, we present the crystal structures of the evolved Stoffel fragment of Taq DNA polymerase SFM4-3 processing the C2'-OMe-GTP in different states. Our results reveal the structural basis for recognition of C2'-methoxy by SFM4-3. Based on the analysis of other mutated residues in SFM4-3, a new Stoffel fragment variant with faster catalytic rate and stronger inhibitor-resistance was obtained. In addition, the capture of a novel pre-insertion co-existing with template 5'-overhang stacking conformation provides insight into the catalytic mechanism of Taq DNA polymerase.


Subject(s)
Models, Molecular , Crystallography, X-Ray , Protein Conformation , DNA/metabolism , DNA/chemistry , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Taq Polymerase/metabolism , Taq Polymerase/chemistry
17.
Am J Hum Genet ; 111(9): 1970-1993, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39106866

ABSTRACT

The precise regulation of DNA replication is vital for cellular division and genomic integrity. Central to this process is the replication factor C (RFC) complex, encompassing five subunits, which loads proliferating cell nuclear antigen onto DNA to facilitate the recruitment of replication and repair proteins and enhance DNA polymerase processivity. While RFC1's role in cerebellar ataxia, neuropathy, and vestibular areflexia syndrome (CANVAS) is known, the contributions of RFC2-5 subunits on human Mendelian disorders is largely unexplored. Our research links bi-allelic variants in RFC4, encoding a core RFC complex subunit, to an undiagnosed disorder characterized by incoordination and muscle weakness, hearing impairment, and decreased body weight. We discovered across nine affected individuals rare, conserved, predicted pathogenic variants in RFC4, all likely to disrupt the C-terminal domain indispensable for RFC complex formation. Analysis of a previously determined cryo-EM structure of RFC bound to proliferating cell nuclear antigen suggested that the variants disrupt interactions within RFC4 and/or destabilize the RFC complex. Cellular studies using RFC4-deficient HeLa cells and primary fibroblasts demonstrated decreased RFC4 protein, compromised stability of the other RFC complex subunits, and perturbed RFC complex formation. Additionally, functional studies of the RFC4 variants affirmed diminished RFC complex formation, and cell cycle studies suggested perturbation of DNA replication and cell cycle progression. Our integrated approach of combining in silico, structural, cellular, and functional analyses establishes compelling evidence that bi-allelic loss-of-function RFC4 variants contribute to the pathogenesis of this multisystemic disorder. These insights broaden our understanding of the RFC complex and its role in human health and disease.


Subject(s)
Replication Protein C , Humans , Replication Protein C/genetics , Replication Protein C/metabolism , Male , HeLa Cells , Female , Phenotype , DNA Replication/genetics , Adult , Mutation , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/genetics , Alleles
18.
DNA Repair (Amst) ; 141: 103740, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39096696

ABSTRACT

An organism's genomic DNA must be accurately duplicated during each cell cycle. DNA synthesis is catalysed by DNA polymerase enzymes, which extend nucleotide polymers in a 5' to 3' direction. This inherent directionality necessitates that one strand is synthesised forwards (leading), while the other is synthesised backwards discontinuously (lagging) to couple synthesis to the unwinding of duplex DNA. Eukaryotic cells possess many diverse polymerases that coordinate to replicate DNA, with the three main replicative polymerases being Pol α, Pol δ and Pol ε. Studies conducted in yeasts and human cells utilising mutant polymerases that incorporate molecular signatures into nascent DNA implicate Pol ε in leading strand synthesis and Pol α and Pol δ in lagging strand replication. Recent structural insights have revealed how the spatial organization of these enzymes around the core helicase facilitates their strand-specific roles. However, various challenging situations during replication require flexibility in the usage of these enzymes, such as during replication initiation or encounters with replication-blocking adducts. This review summarises the roles of the replicative polymerases in bulk DNA replication and explores their flexible and dynamic deployment to complete genome replication. We also examine how polymerase usage patterns can inform our understanding of global replication dynamics by revealing replication fork directionality to identify regions of replication initiation and termination.


Subject(s)
DNA Replication , Humans , DNA/metabolism , DNA/biosynthesis , DNA-Directed DNA Polymerase/metabolism , Animals , DNA Polymerase II/metabolism , Eukaryota/enzymology , Eukaryota/genetics , DNA Polymerase III/metabolism , Eukaryotic Cells/metabolism , Eukaryotic Cells/enzymology , DNA Polymerase I/metabolism
19.
Genomics ; 116(5): 110908, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39106913

ABSTRACT

DNA replication is a fundamental process for cell proliferation, governed by intricate mechanisms involving leading and lagging strand synthesis. In eukaryotes, canonical DNA replication occurs during the S phase of the cell cycle, facilitated by various components of the replicative machinery at sites known as replication origins. Leading and lagging strands exhibit distinct replication dynamics, with leading strand replication being relatively straightforward compared to the complex synthesis of lagging strands involving Okazaki fragment maturation. Central to DNA synthesis are DNA polymerases, with Polα, Polε, and Polδ playing pivotal roles, each specializing in specific tasks during replication. Notably, leading and lagging strands are replicated by different polymerases, contributing to the division of labor in DNA replication. Understanding the enzymology of asymmetric DNA replication has been challenging, with methods relying on ribonucleotide incorporation and next-generation sequencing techniques offering comprehensive insights. These methodologies, such as HydEn-seq, PU-seq, ribose-seq, and emRiboSeq, offer insights into polymerase activity and strand synthesis, aiding in understanding DNA replication dynamics. Recent advancements include novel conditional mutants for ribonucleotide excision repair, enzymatic cleavage alternatives, and unified pipelines for data analysis. Further developments in adapting techniques to different organisms, studying non-canonical polymerases, and exploring new sequencing platforms hold promise for expanding our understanding of DNA replication dynamics. Integrating strand-specific information into single-cell studies could offer novel insights into enzymology, opening avenues for future research and applications in repair and replication biology.

20.
DNA Repair (Amst) ; 141: 103731, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089193

ABSTRACT

DNA replication is remarkably accurate with estimates of only a handful of mutations per human genome per cell division cycle. Replication stress caused by DNA lesions, transcription-replication conflicts, and other obstacles to the replication machinery must be efficiently overcome in ways that minimize errors and maximize completion of DNA synthesis. Replication fork reversal is one mechanism that helps cells tolerate replication stress. This process involves reannealing of parental template DNA strands and generation of a nascent-nascent DNA duplex. While fork reversal may be beneficial by facilitating DNA repair or template switching, it must be confined to the appropriate contexts to preserve genome stability. Many enzymes have been implicated in this process including ATP-dependent DNA translocases like SMARCAL1, ZRANB3, HLTF, and the helicase FBH1. In addition, the RAD51 recombinase is required. Many additional factors and regulatory activities also act to ensure reversal is beneficial instead of yielding undesirable outcomes. Finally, reversed forks must also be stabilized and often need to be restarted to complete DNA synthesis. Disruption or deregulation of fork reversal causes a variety of human diseases. In this review we will describe the latest models for reversal and key mechanisms of regulation.


Subject(s)
DNA Damage Tolerance , Animals , Humans , DNA/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , DNA Repair , Genomic Instability
SELECTION OF CITATIONS
SEARCH DETAIL