Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.358
Filter
Add more filters

Publication year range
1.
Cell ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39197451

ABSTRACT

DNA polymerases are important drug targets, and many structural studies have captured them in distinct conformations. However, a detailed understanding of the impact of polymerase conformational dynamics on drug resistance is lacking. We determined cryoelectron microscopy (cryo-EM) structures of DNA-bound herpes simplex virus polymerase holoenzyme in multiple conformations and interacting with antivirals in clinical use. These structures reveal how the catalytic subunit Pol and the processivity factor UL42 bind DNA to promote processive DNA synthesis. Unexpectedly, in the absence of an incoming nucleotide, we observed Pol in multiple conformations with the closed state sampled by the fingers domain. Drug-bound structures reveal how antivirals may selectively bind enzymes that more readily adopt the closed conformation. Molecular dynamics simulations and the cryo-EM structure of a drug-resistant mutant indicate that some resistance mutations modulate conformational dynamics rather than directly impacting drug binding, thus clarifying mechanisms that drive drug selectivity.

2.
Annu Rev Biochem ; 91: 133-155, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35287470

ABSTRACT

Our current view of how DNA-based genomes are efficiently and accurately replicated continues to evolve as new details emerge on the presence of ribonucleotides in DNA. Ribonucleotides are incorporated during eukaryotic DNA replication at rates that make them the most common noncanonical nucleotide placed into the nuclear genome, they are efficiently repaired, and their removal impacts genome integrity. This review focuses on three aspects of this subject: the incorporation of ribonucleotides into the eukaryotic nuclear genome during replication by B-family DNA replicases, how these ribonucleotides are removed, and the consequences of their presence or removal for genome stability and disease.


Subject(s)
DNA Replication , Genomic Instability , Ribonucleotides , DNA/genetics , DNA/metabolism , DNA Repair , Eukaryota/genetics , Eukaryota/metabolism , Nucleotidyltransferases/genetics , Ribonucleotides/genetics , Ribonucleotides/metabolism
3.
Annu Rev Biochem ; 88: 137-162, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31220977

ABSTRACT

Genomic DNA is susceptible to endogenous and environmental stresses that modify DNA structure and its coding potential. Correspondingly, cells have evolved intricate DNA repair systems to deter changes to their genetic material. Base excision DNA repair involves a number of enzymes and protein cofactors that hasten repair of damaged DNA bases. Recent advances have identified macromolecular complexes that assemble at the DNA lesion and mediate repair. The repair of base lesions generally requires five enzymatic activities: glycosylase, endonuclease, lyase, polymerase, and ligase. The protein cofactors and mechanisms for coordinating the sequential enzymatic steps of repair are being revealed through a range of experimental approaches. We discuss the enzymes and protein cofactors involved in eukaryotic base excision repair, emphasizing the challenge of integrating findings from multiple methodologies. The results provide an opportunity to assimilate biochemical findings with cell-based assays to uncover new insights into this deceptively complex repair pathway.


Subject(s)
DNA Glycosylases/chemistry , DNA-Directed DNA Polymerase/chemistry , DNA/chemistry , Endonucleases/chemistry , Genome , Ligases/chemistry , Lyases/chemistry , DNA/metabolism , DNA/ultrastructure , DNA Damage , DNA Glycosylases/metabolism , DNA Glycosylases/ultrastructure , DNA Repair , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/ultrastructure , Endonucleases/metabolism , Endonucleases/ultrastructure , Eukaryota/genetics , Eukaryota/metabolism , Eukaryotic Cells/cytology , Eukaryotic Cells/enzymology , Genomic Instability , Humans , Ligases/metabolism , Ligases/ultrastructure , Lyases/metabolism , Lyases/ultrastructure , Models, Molecular , Mutagenesis , Nucleic Acid Conformation , Protein Conformation
4.
Annu Rev Biochem ; 88: 163-190, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31220976

ABSTRACT

Many DNA-processing enzymes have been shown to contain a [4Fe4S] cluster, a common redox cofactor in biology. Using DNA electrochemistry, we find that binding of the DNA polyanion promotes a negative shift in [4Fe4S] cluster potential, which corresponds thermodynamically to a ∼500-fold increase in DNA-binding affinity for the oxidized [4Fe4S]3+ cluster versus the reduced [4Fe4S]2+ cluster. This redox switch can be activated from a distance using DNA charge transport (DNA CT) chemistry. DNA-processing proteins containing the [4Fe4S] cluster are enumerated, with possible roles for the redox switch highlighted. A model is described where repair proteins may signal one another using DNA-mediated charge transport as a first step in their search for lesions. The redox switch in eukaryotic DNA primases appears to regulate polymerase handoff, and in DNA polymerase δ, the redox switch provides a means to modulate replication in response to oxidative stress. We thus describe redox signaling interactions of DNA-processing [4Fe4S] enzymes, as well as the most interesting potential players to consider in delineating new DNA-mediated redox signaling networks.


Subject(s)
DNA Glycosylases/chemistry , DNA Helicases/chemistry , DNA-Directed DNA Polymerase/chemistry , DNA/chemistry , Endonucleases/chemistry , Genome , Iron-Sulfur Proteins/chemistry , Animals , Bacteria/genetics , Bacteria/metabolism , DNA/metabolism , DNA/ultrastructure , DNA Damage , DNA Glycosylases/metabolism , DNA Glycosylases/ultrastructure , DNA Helicases/metabolism , DNA Helicases/ultrastructure , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/ultrastructure , Electron Spin Resonance Spectroscopy , Endonucleases/metabolism , Endonucleases/ultrastructure , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/ultrastructure , Oxidation-Reduction , Protein Binding , Signal Transduction , Thermodynamics
5.
Cell ; 176(6): 1295-1309.e15, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30773314

ABSTRACT

Cancers from sun-exposed skin accumulate "driver" mutations, causally implicated in oncogenesis. Because errors incorporated during translesion synthesis (TLS) opposite UV lesions would generate these mutations, TLS mechanisms are presumed to underlie cancer development. To address the role of TLS in skin cancer formation, we determined which DNA polymerase is responsible for generating UV mutations, analyzed the relative contributions of error-free TLS by Polη and error-prone TLS by Polθ to the replication of UV-damaged DNA and to genome stability, and examined the incidence of UV-induced skin cancers in Polθ-/-, Polη-/-, and Polθ-/- Polη-/- mice. Our findings that the incidence of skin cancers rises in Polθ-/- mice and is further exacerbated in Polθ-/- Polη-/- mice compared with Polη-/- mice support the conclusion that error-prone TLS by Polθ provides a safeguard against tumorigenesis and suggest that cancer formation can ensue in the absence of somatic point mutations.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/physiology , Skin Neoplasms/metabolism , Animals , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/physiology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Genomic Instability/genetics , Humans , Mice , Mice, Knockout , Mutation/genetics , Skin/cytology , Skin/metabolism , Skin Neoplasms/genetics , Ultraviolet Rays/adverse effects , DNA Polymerase theta
6.
Annu Rev Biochem ; 86: 417-438, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28301743

ABSTRACT

This review focuses on the biogenesis and composition of the eukaryotic DNA replication fork, with an emphasis on the enzymes that synthesize DNA and repair discontinuities on the lagging strand of the replication fork. Physical and genetic methodologies aimed at understanding these processes are discussed. The preponderance of evidence supports a model in which DNA polymerase ε (Pol ε) carries out the bulk of leading strand DNA synthesis at an undisturbed replication fork. DNA polymerases α and δ carry out the initiation of Okazaki fragment synthesis and its elongation and maturation, respectively. This review also discusses alternative proposals, including cellular processes during which alternative forks may be utilized, and new biochemical studies with purified proteins that are aimed at reconstituting leading and lagging strand DNA synthesis separately and as an integrated replication fork.


Subject(s)
DNA Helicases/genetics , DNA Polymerase II/genetics , DNA Replication , DNA/genetics , Eukaryotic Cells/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA/metabolism , DNA Helicases/metabolism , DNA Polymerase I/genetics , DNA Polymerase I/metabolism , DNA Polymerase II/metabolism , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Eukaryotic Cells/cytology , Humans , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/metabolism
7.
Cell ; 169(7): 1201-1213.e17, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28622507

ABSTRACT

It has been assumed that DNA synthesis by the leading- and lagging-strand polymerases in the replisome must be coordinated to avoid the formation of significant gaps in the nascent strands. Using real-time single-molecule analysis, we establish that leading- and lagging-strand DNA polymerases function independently within a single replisome. Although average rates of DNA synthesis on leading and lagging strands are similar, individual trajectories of both DNA polymerases display stochastically switchable rates of synthesis interspersed with distinct pauses. DNA unwinding by the replicative helicase may continue during such pauses, but a self-governing mechanism, where helicase speed is reduced by ∼80%, permits recoupling of polymerase to helicase. These features imply a more dynamic, kinetically discontinuous replication process, wherein contacts within the replisome are continually broken and reformed. We conclude that the stochastic behavior of replisome components ensures complete DNA duplication without requiring coordination of leading- and lagging-strand synthesis. PAPERCLIP.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase/metabolism , Escherichia coli/metabolism , DNA Helicases/metabolism , Escherichia coli/enzymology , Microscopy, Fluorescence/methods , Models, Biological , Replicon
8.
Mol Cell ; 84(8): 1460-1474.e6, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38640894

ABSTRACT

DNA polymerase θ (Polθ) plays a central role in a DNA double-strand break repair pathway termed theta-mediated end joining (TMEJ). TMEJ functions by pairing short-sequence "microhomologies" (MHs) in single-stranded DNA at each end of a break and subsequently initiating DNA synthesis. It is not known how the Polθ helicase domain (HD) and polymerase domain (PD) operate to bring together MHs and facilitate repair. To resolve these transient processes in real time, we utilized in vitro single-molecule FRET approaches and biochemical analyses. We find that the Polθ-HD mediates the initial capture of two ssDNA strands, bringing them in close proximity. The Polθ-PD binds and stabilizes pre-annealed MHs to form a synaptic complex (SC) and initiate repair synthesis. Individual synthesis reactions show that Polθ is inherently non-processive, accounting for complex mutational patterns during TMEJ. Binding of Polθ-PD to stem-loop-forming sequences can substantially limit synapsis, depending on the available dNTPs and sequence context.


Subject(s)
DNA Breaks, Double-Stranded , DNA-Directed DNA Polymerase , DNA-Directed DNA Polymerase/metabolism , DNA Replication , DNA, Single-Stranded/genetics , DNA Helicases/genetics , DNA End-Joining Repair
9.
Mol Cell ; 83(16): 2911-2924.e16, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37506699

ABSTRACT

During eukaryotic DNA replication, Pol α-primase generates primers at replication origins to start leading-strand synthesis and every few hundred nucleotides during discontinuous lagging-strand replication. How Pol α-primase is targeted to replication forks to prime DNA synthesis is not fully understood. Here, by determining cryoelectron microscopy (cryo-EM) structures of budding yeast and human replisomes containing Pol α-primase, we reveal a conserved mechanism for the coordination of priming by the replisome. Pol α-primase binds directly to the leading edge of the CMG (CDC45-MCM-GINS) replicative helicase via a complex interaction network. The non-catalytic PRIM2/Pri2 subunit forms two interfaces with CMG that are critical for in vitro DNA replication and yeast cell growth. These interactions position the primase catalytic subunit PRIM1/Pri1 directly above the exit channel for lagging-strand template single-stranded DNA (ssDNA), revealing why priming occurs efficiently only on the lagging-strand template and elucidating a mechanism for Pol α-primase to overcome competition from RPA to initiate primer synthesis.


Subject(s)
DNA Primase , DNA Replication , Humans , DNA Primase/genetics , DNA Primase/metabolism , Cryoelectron Microscopy , DNA Helicases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , DNA, Single-Stranded/metabolism
10.
Mol Cell ; 81(7): 1534-1547.e4, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33577776

ABSTRACT

Cancers with hereditary defects in homologous recombination rely on DNA polymerase θ (pol θ) for repair of DNA double-strand breaks. During end joining, pol θ aligns microhomology tracts internal to 5'-resected broken ends. An unidentified nuclease trims the 3' ends before synthesis can occur. Here we report that a nuclease activity, which differs from the proofreading activity often associated with DNA polymerases, is intrinsic to the polymerase domain of pol θ. Like the DNA synthesis activity, the nuclease activity requires conserved metal-binding residues, metal ions, and dNTPs and is inhibited by ddNTPs or chain-terminated DNA. Our data indicate that pol θ repurposes metal ions in the polymerase active site for endonucleolytic cleavage and that the polymerase-active and end-trimming conformations of the enzyme are distinct. We reveal a nimble strategy of substrate processing that allows pol θ to trim or extend DNA depending on the DNA repair context.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Directed DNA Polymerase/metabolism , DNA/metabolism , Endonucleases/metabolism , Metals/metabolism , Cell Line , DNA/genetics , DNA-Directed DNA Polymerase/genetics , Endonucleases/genetics , Humans , DNA Polymerase theta
11.
Mol Cell ; 81(13): 2778-2792.e4, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33932350

ABSTRACT

DNA polymerase ε (Polε) carries out high-fidelity leading strand synthesis owing to its exonuclease activity. Polε polymerase and exonuclease activities are balanced, because of partitioning of nascent DNA strands between catalytic sites, so that net resection occurs when synthesis is impaired. In vivo, DNA synthesis stalling activates replication checkpoint kinases, which act to preserve the functional integrity of replication forks. We show that stalled Polε drives nascent strand resection causing fork functional collapse, averted via checkpoint-dependent phosphorylation. Polε catalytic subunit Pol2 is phosphorylated on serine 430, influencing partitioning between polymerase and exonuclease active sites. A phosphormimetic S430D change reduces exonucleolysis in vitro and counteracts fork collapse. Conversely, non-phosphorylatable pol2-S430A expression causes resection-driven stressed fork defects. Our findings reveal that checkpoint kinases switch Polε to an exonuclease-safe mode preventing nascent strand resection and stabilizing stalled replication forks. Elective partitioning suppression has implications for the diverse Polε roles in genome integrity maintenance.


Subject(s)
DNA Polymerase II/chemistry , Exonucleases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Amino Acid Substitution , Catalytic Domain , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , DNA, Fungal/biosynthesis , DNA, Fungal/chemistry , DNA, Fungal/genetics , Exonucleases/genetics , Exonucleases/metabolism , Mutation, Missense , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
12.
EMBO J ; 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039289

ABSTRACT

Unintended on-target chromosomal alterations induced by CRISPR/Cas9 in mammalian cells are common, particularly large deletions and chromosomal translocations, and present a safety challenge for genome editing. Thus, there is still an unmet need to develop safer and more efficient editing tools. We screened diverse DNA polymerases of distinct origins and identified a T4 DNA polymerase derived from phage T4 that strongly prevents undesired on-target damage while increasing the proportion of precise 1- to 2-base-pair insertions generated during CRISPR/Cas9 editing (termed CasPlus). CasPlus induced substantially fewer on-target large deletions while increasing the efficiency of correcting common frameshift mutations in DMD and restored higher level of dystrophin expression than Cas9-alone in human cardiomyocytes. Moreover, CasPlus greatly reduced the frequency of on-target large deletions during mouse germline editing. In multiplexed guide RNAs mediating gene editing, CasPlus repressed chromosomal translocations while maintaining gene disruption efficiency that was higher or comparable to Cas9 in primary human T cells. Therefore, CasPlus offers a safer and more efficient gene editing strategy to treat pathogenic variants or to introduce genetic modifications in human applications.

13.
Mol Cell ; 78(6): 1166-1177.e6, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32497495

ABSTRACT

Human tumors with exonuclease domain mutations in the gene encoding DNA polymerase ε (POLE) have incredibly high mutation burdens. These errors arise in four unique mutation signatures occurring in different relative amounts, the etiologies of which remain poorly understood. We used CRISPR-Cas9 to engineer human cell lines expressing POLE tumor variants, with and without mismatch repair (MMR). Whole-exome sequencing of these cells after defined numbers of population doublings permitted analysis of nascent mutation accumulation. Unlike an exonuclease active site mutant that we previously characterized, POLE cancer mutants readily drive signature mutagenesis in the presence of functional MMR. Comparison of cell line and human patient data suggests that the relative abundance of mutation signatures partitions POLE tumors into distinct subgroups dependent on the nature of the POLE allele, its expression level, and MMR status. These results suggest that different POLE mutants have previously unappreciated differences in replication fidelity and mutagenesis.


Subject(s)
DNA Mismatch Repair/genetics , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , Alleles , Cell Line, Tumor , DNA Mismatch Repair/physiology , Humans , Mutagenesis/genetics , Mutation/genetics , Neoplasms/genetics , Neoplasms/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism
14.
Mol Cell ; 80(1): 114-126.e8, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32916094

ABSTRACT

DNA replication is carried out by a multi-protein machine called the replisome. In Saccharomyces cerevisiae, the replisome is composed of over 30 different proteins arranged into multiple subassemblies, each performing distinct activities. Synchrony of these activities is required for efficient replication and preservation of genomic integrity. How this is achieved is particularly puzzling at the lagging strand, where current models of the replisome architecture propose turnover of the canonical lagging strand polymerase, Pol δ, at every cycle of Okazaki fragment synthesis. Here, we established single-molecule fluorescence microscopy protocols to study the binding kinetics of individual replisome subunits in live S. cerevisiae. Our results show long residence times for most subunits at the active replisome, supporting a model where all subassemblies bind tightly and work in a coordinated manner for extended periods, including Pol δ, redefining the architecture of the active eukaryotic replisome.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase/metabolism , Eukaryotic Cells/metabolism , Multienzyme Complexes/metabolism , Cell Nucleus/metabolism , Kinetics , Models, Biological , Nuclear Proteins/metabolism , Protein Subunits/metabolism , Reproducibility of Results , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Single Molecule Imaging , Time Factors
15.
Mol Cell ; 79(6): 1037-1050.e5, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32882183

ABSTRACT

DNA double-stranded breaks (DSBs) are dangerous lesions threatening genomic stability. Fidelity of DSB repair is best achieved by recombination with a homologous template sequence. In yeast, transcript RNA was shown to template DSB repair of DNA. However, molecular pathways of RNA-driven repair processes remain obscure. Utilizing assays of RNA-DNA recombination with and without an induced DSB in yeast DNA, we characterize three forms of RNA-mediated genomic modifications: RNA- and cDNA-templated DSB repair (R-TDR and c-TDR) using an RNA transcript or a DNA copy of the RNA transcript for DSB repair, respectively, and a new mechanism of RNA-templated DNA modification (R-TDM) induced by spontaneous or mutagen-induced breaks. While c-TDR requires reverse transcriptase, translesion DNA polymerase ζ (Pol ζ) plays a major role in R-TDR, and it is essential for R-TDM. This study characterizes mechanisms of RNA-DNA recombination, uncovering a role of Pol ζ in transferring genetic information from transcript RNA to DNA.


Subject(s)
DNA/genetics , RNA/genetics , Saccharomyces cerevisiae/genetics , Adolescent , Adult , DNA/ultrastructure , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA Replication/genetics , DNA, Complementary/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/ultrastructure , Genomic Instability/genetics , Humans , Middle Aged , RNA/ultrastructure , Rad52 DNA Repair and Recombination Protein/genetics , Young Adult
16.
Trends Biochem Sci ; 48(10): 860-872, 2023 10.
Article in English | MEDLINE | ID: mdl-37586999

ABSTRACT

Telomere maintenance is essential for the genome integrity of eukaryotes, and this function is underpinned by the two-step telomeric DNA synthesis process: telomere G-overhang extension by telomerase and complementary strand fill-in by DNA polymerase alpha-primase (polα-primase). Compared to the telomerase step, the telomere C-strand fill-in mechanism is less understood. Recent studies have provided new insights into how telomeric single-stranded DNA-binding protein CTC1-STN1-TEN1 (CST) and polα-primase coordinate to synthesize the telomeric C-strand for telomere overhang fill-in. Cryogenic electron microscopy (cryo-EM) structures of CST-polα-primase complexes have provided additional insights into how they assemble at telomeric templates and de novo synthesize the telomere C-strand. In this review, we discuss how these latest findings coalesce with existing understanding to develop a human telomere C-strand fill-in mechanism model.


Subject(s)
DNA Primase , Telomerase , Humans , Telomere , Shelterin Complex , Eukaryota
17.
Mol Cell ; 76(3): 371-381.e4, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31495565

ABSTRACT

Break-induced replication (BIR) is a pathway of homology-directed repair that repairs one-ended DNA breaks, such as those formed at broken replication forks or uncapped telomeres. In contrast to conventional S phase DNA synthesis, BIR proceeds by a migrating D-loop and results in conservative synthesis of the nascent strands. DNA polymerase delta (Pol δ) initiates BIR; however, it is not known whether synthesis of the invading strand switches to a different polymerase or how the complementary strand is synthesized. By using alleles of the replicative DNA polymerases that are permissive for ribonucleotide incorporation, thus generating a signature of their action in the genome that can be identified by hydrolytic end sequencing, we show that Pol δ replicates both the invading and the complementary strand during BIR. In support of this conclusion, we show that depletion of Pol δ from cells reduces BIR, whereas depletion of Pol ε has no effect.


Subject(s)
DNA Breaks , DNA Polymerase III/metabolism , DNA Replication , DNA, Fungal/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , DNA Ligase ATP/genetics , DNA Ligase ATP/metabolism , DNA Polymerase I/genetics , DNA Polymerase I/metabolism , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , DNA Polymerase III/genetics , DNA, Fungal/genetics , HEK293 Cells , HeLa Cells , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
18.
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
19.
Proc Natl Acad Sci U S A ; 121(20): e2400610121, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38713623

ABSTRACT

Chromatin replication is intricately intertwined with the recycling of parental histones to the newly duplicated DNA strands for faithful genetic and epigenetic inheritance. The transfer of parental histones occurs through two distinct pathways: leading strand deposition, mediated by the DNA polymerase ε subunits Dpb3/Dpb4, and lagging strand deposition, facilitated by the MCM helicase subunit Mcm2. However, the mechanism of the facilitation of Mcm2 transferring parental histones to the lagging strand while moving along the leading strand remains unclear. Here, we show that the deletion of Pol32, a nonessential subunit of major lagging-strand DNA polymerase δ, results in a predominant transfer of parental histone H3-H4 to the leading strand during replication. Biochemical analyses further demonstrate that Pol32 can bind histone H3-H4 both in vivo and in vitro. The interaction of Pol32 with parental histone H3-H4 is disrupted through the mutation of the histone H3-H4 binding domain within Mcm2. Our findings identify the DNA polymerase δ subunit Pol32 as a critical histone chaperone downstream of Mcm2, mediating the transfer of parental histones to the lagging strand during DNA replication.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase , Saccharomyces cerevisiae Proteins , DNA Polymerase III/metabolism , DNA Polymerase III/genetics , Histones/metabolism , Minichromosome Maintenance Complex Component 2/metabolism , Minichromosome Maintenance Complex Component 2/genetics , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , DNA-Directed DNA Polymerase/metabolism
20.
Proc Natl Acad Sci U S A ; 121(23): e2405771121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38805295

ABSTRACT

The phylum Preplasmiviricota (kingdom Bamfordvirae, realm Varidnaviria) is a broad assemblage of diverse viruses with comparatively short double-stranded DNA genomes (<50 kbp) that produce icosahedral capsids built from double jelly-roll major capsid proteins. Preplasmiviricots infect hosts from all cellular domains, testifying to their ancient origin, and, in particular, are associated with six of the seven supergroups of eukaryotes. Preplasmiviricots comprise four major groups of viruses, namely, polintons, polinton-like viruses (PLVs), virophages, and adenovirids. We used protein structure modeling and analysis to show that protein-primed DNA polymerases (pPolBs) of polintons, virophages, and cytoplasmic linear plasmids encompass an N-terminal domain homologous to the terminal proteins (TPs) of prokaryotic PRD1-like tectivirids and eukaryotic adenovirids that are involved in protein-primed replication initiation, followed by a viral ovarian tumor-like cysteine deubiquitinylase (vOTU) domain. The vOTU domain is likely responsible for the cleavage of the TP from the large pPolB polypeptide and is inactivated in adenovirids, in which TP is a separate protein. Many PLVs and transpovirons encode a distinct derivative of polinton-like pPolB that retains the TP, vOTU, and pPolB polymerization palm domains but lacks the exonuclease domain and instead contains a superfamily 1 helicase domain. Analysis of the presence/absence and inactivation of the vOTU domains and replacement of pPolB with other DNA polymerases in eukaryotic preplasmiviricots enabled us to outline a complete scenario for their origin and evolution.


Subject(s)
Capsid Proteins , DNA Viruses , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , DNA Viruses/genetics , Eukaryota/virology , Eukaryota/genetics , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , Models, Molecular , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL