Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 291
Filtrar
1.
Nature ; 629(8011): 467-473, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38471529

RESUMEN

Prokaryotes have evolved intricate innate immune systems against phage infection1-7. Gabija is a highly widespread prokaryotic defence system that consists of two components, GajA and GajB8. GajA functions as a DNA endonuclease that is inactive in the presence of ATP9. Here, to explore how the Gabija system is activated for anti-phage defence, we report its cryo-electron microscopy structures in five states, including apo GajA, GajA in complex with DNA, GajA bound by ATP, apo GajA-GajB, and GajA-GajB in complex with ATP and Mg2+. GajA is a rhombus-shaped tetramer with its ATPase domain clustered at the centre and the topoisomerase-primase (Toprim) domain located peripherally. ATP binding at the ATPase domain stabilizes the insertion region within the ATPase domain, keeping the Toprim domain in a closed state. Upon ATP depletion by phages, the Toprim domain opens to bind and cleave the DNA substrate. GajB, which docks on GajA, is activated by the cleaved DNA, ultimately leading to prokaryotic cell death. Our study presents a mechanistic landscape of Gabija activation.


Asunto(s)
Bacillus cereus , Proteínas Bacterianas , Bacteriófagos , Microscopía por Crioelectrón , Inmunidad Innata , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/ultraestructura , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Apoproteínas/química , Apoproteínas/inmunología , Apoproteínas/metabolismo , Apoproteínas/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Bacteriófagos/inmunología , ADN/metabolismo , ADN/química , División del ADN , Magnesio/química , Magnesio/metabolismo , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Viabilidad Microbiana , Bacillus cereus/química , Bacillus cereus/inmunología , Bacillus cereus/metabolismo , Bacillus cereus/ultraestructura , Estructura Cuaternaria de Proteína , ADN Primasa/química , ADN Primasa/metabolismo , ADN Primasa/ultraestructura , ADN-Topoisomerasas/química , ADN-Topoisomerasas/metabolismo , ADN-Topoisomerasas/ultraestructura
2.
Nature ; 608(7924): 826-832, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35830881

RESUMEN

The mammalian DNA polymerase-α-primase (Polα-primase) complex is essential for DNA metabolism, providing the de novo RNA-DNA primer for several DNA replication pathways1-4 such as lagging-strand synthesis and telomere C-strand fill-in. The physical mechanism underlying how Polα-primase, alone or in partnership with accessory proteins, performs its complicated multistep primer synthesis function is unknown. Here we show that CST, a single-stranded DNA-binding accessory protein complex for Polα-primase, physically organizes the enzyme for efficient primer synthesis. Cryogenic electron microscopy structures of the CST-Polα-primase preinitiation complex (PIC) bound to various types of telomere overhang reveal that template-bound CST partitions the DNA and RNA catalytic centres of Polα-primase into two separate domains and effectively arranges them in RNA-DNA synthesis order. The architecture of the PIC provides a single solution for the multiple structural requirements for the synthesis of RNA-DNA primers by Polα-primase. Several insights into the template-binding specificity of CST, template requirement for assembly of the CST-Polα-primase PIC and activation are also revealed in this study.


Asunto(s)
ADN Primasa , Complejo Shelterina , Telómero , Moldes Genéticos , ADN/metabolismo , ADN Primasa/química , ADN Primasa/metabolismo , Cartilla de ADN/biosíntesis , Replicación del ADN , Humanos , Dominios Proteicos , ARN/biosíntesis , ARN/metabolismo , Complejo Shelterina/química , Complejo Shelterina/metabolismo , Especificidad por Sustrato , Telómero/química , Telómero/genética , Telómero/metabolismo
3.
Nature ; 608(7924): 813-818, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35831498

RESUMEN

Telomeres are the physical ends of linear chromosomes. They are composed of short repeating sequences (such as TTGGGG in the G-strand for Tetrahymena thermophila) of double-stranded DNA with a single-strand 3' overhang of the G-strand and, in humans, the six shelterin proteins: TPP1, POT1, TRF1, TRF2, RAP1 and TIN21,2. TPP1 and POT1 associate with the 3' overhang, with POT1 binding the G-strand3 and TPP1 (in complex with TIN24) recruiting telomerase via interaction with telomerase reverse transcriptase5 (TERT). The telomere DNA ends are replicated and maintained by telomerase6, for the G-strand, and subsequently DNA polymerase α-primase7,8 (PolαPrim), for the C-strand9. PolαPrim activity is stimulated by the heterotrimeric complex CTC1-STN1-TEN110-12 (CST), but the structural basis of the recruitment of PolαPrim and CST to telomere ends remains unknown. Here we report cryo-electron microscopy (cryo-EM) structures of Tetrahymena CST in the context of the telomerase holoenzyme, in both the absence and the presence of PolαPrim, and of PolαPrim alone. Tetrahymena Ctc1 binds telomerase subunit p50, a TPP1 orthologue, on a flexible Ctc1 binding motif revealed by cryo-EM and NMR spectroscopy. The PolαPrim polymerase subunit POLA1 binds Ctc1 and Stn1, and its interface with Ctc1 forms an entry port for G-strand DNA to the POLA1 active site. We thus provide a snapshot of four key components that are required for telomeric DNA synthesis in a single active complex-telomerase-core ribonucleoprotein, p50, CST and PolαPrim-that provides insights into the recruitment of CST and PolαPrim and the handoff between G-strand and C-strand synthesis.


Asunto(s)
ADN Primasa , Complejo Shelterina , Telomerasa , Tetrahymena , Microscopía por Crioelectrón , ADN/genética , ADN/metabolismo , ADN Primasa/química , ADN Primasa/metabolismo , ADN Primasa/ultraestructura , Holoenzimas/química , Holoenzimas/metabolismo , Holoenzimas/ultraestructura , Unión Proteica , Complejo Shelterina/química , Complejo Shelterina/metabolismo , Complejo Shelterina/ultraestructura , Telomerasa/química , Telomerasa/metabolismo , Telomerasa/ultraestructura , Telómero/genética , Telómero/metabolismo , Tetrahymena/química , Tetrahymena/enzimología , Tetrahymena/metabolismo , Tetrahymena/ultraestructura
4.
Genes Dev ; 34(21-22): 1520-1533, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33060134

RESUMEN

DNA replication is fundamental for cell proliferation in all organisms. Nonetheless, components of the replisome have been implicated in human disease, and here we report PRIM1 encoding the catalytic subunit of DNA primase as a novel disease gene. Using a variant classification agnostic approach, biallelic mutations in PRIM1 were identified in five individuals. PRIM1 protein levels were markedly reduced in patient cells, accompanied by replication fork asymmetry, increased interorigin distances, replication stress, and prolonged S-phase duration. Consequently, cell proliferation was markedly impaired, explaining the patients' extreme growth failure. Notably, phenotypic features distinct from those previously reported with DNA polymerase genes were evident, highlighting differing developmental requirements for this core replisome component that warrant future investigation.


Asunto(s)
ADN Primasa/genética , Enanismo/genética , Retardo del Crecimiento Fetal/genética , ADN Primasa/química , ADN Primasa/deficiencia , Enanismo/diagnóstico por imagen , Enanismo/patología , Femenino , Retardo del Crecimiento Fetal/diagnóstico por imagen , Retardo del Crecimiento Fetal/patología , Variación Genética , Humanos , Lactante , Masculino , Linaje , Síndrome
5.
Nucleic Acids Res ; 52(9): 4818-4829, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38597656

RESUMEN

Protein binding microarrays (PBM), SELEX, RNAcompete and chromatin-immunoprecipitation have been intensively used to determine the specificity of nucleic acid binding proteins. While the specificity of proteins with pronounced sequence specificity is straightforward, the determination of the sequence specificity of proteins of modest sequence specificity is more difficult. In this work, an explorative data analysis workflow for nucleic acid binding data was developed that can be used by scientists that want to analyse their binding data. The workflow is based on a regressor realized in scikit-learn, the major machine learning module for the scripting language Python. The regressor is built on a thermodynamic model of nucleic acid binding and describes the sequence specificity with base- and position-specific energies. The regressor was used to determine the binding specificity of the T7 primase. For this, we reanalysed the binding data of the T7 primase obtained with a custom PBM. The binding specificity of the T7 primase agrees with the priming specificity (5'-GTC) and the template (5'-GGGTC) for the preferentially synthesized tetraribonucleotide primer (5'-pppACCC) but is more relaxed. The dominant contribution of two positions in the motif can be explained by the involvement of the initiating and elongating nucleotides for template binding.


Asunto(s)
Bacteriófago T7 , ADN Primasa , Bacteriófago T7/enzimología , Sitios de Unión , ADN Primasa/metabolismo , ADN Primasa/química , Análisis por Matrices de Proteínas/métodos , Unión Proteica , Termodinámica , Proteínas Virales/metabolismo , Proteínas Virales/química
6.
Subcell Biochem ; 104: 73-100, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38963484

RESUMEN

Telomeres at the end of eukaryotic chromosomes are extended by a specialized set of enzymes and telomere-associated proteins, collectively termed here the telomere "replisome." The telomere replisome acts on a unique replicon at each chromosomal end of the telomeres, the 3' DNA overhang. This telomere replication process is distinct from the replisome mechanism deployed to duplicate the human genome. The G-rich overhang is first extended before the complementary C-strand is filled in. This overhang is extended by telomerase, a specialized ribonucleoprotein and reverse transcriptase. The overhang extension process is terminated when telomerase is displaced by CTC1-STN1-TEN1 (CST), a single-stranded DNA-binding protein complex. CST then recruits DNA polymerase α-primase to complete the telomere replication process by filling in the complementary C-strand. In this chapter, the recent structure-function insights into the human telomere C-strand fill-in machinery (DNA polymerase α-primase and CST) will be discussed.


Asunto(s)
ADN Polimerasa I , ADN Primasa , Replicación del ADN , Proteínas de Unión a Telómeros , Telómero , Humanos , Telómero/metabolismo , Telómero/genética , ADN Polimerasa I/metabolismo , ADN Polimerasa I/genética , ADN Polimerasa I/química , ADN Primasa/metabolismo , ADN Primasa/genética , ADN Primasa/química , Proteínas de Unión a Telómeros/metabolismo , Proteínas de Unión a Telómeros/genética , Telomerasa/metabolismo , Telomerasa/genética
7.
Biophys J ; 123(12): 1648-1653, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38733082

RESUMEN

DNA primase is an iron sulfur enzyme in DNA replication responsible for synthesizing short RNA primers that serve as starting points for DNA synthesis. The role of the [4Fe-4S] cluster is not well determined. Here, we calculate the redox potential of the [4Fe-4S] with and without DNA/RNA using continuum electrostatics. In addition, we identify the structural changes coupled to the oxidation/reduction. Our calculations show that the DNA/RNA primer lowers the redox potential by 110 and 50 mV for the [4Fe-4S]+ and [4Fe-4S]2+ states, respectively. The oxidation of the cluster is coupled to structural changes that significantly reduce the binding energies between the DNA and the nearby residues. The negative charges accumulated by the DNA and the RNA primers induce the oxidation of the [4Fe-4S] cluster. This in turn stimulates structural changes on the DNA-protein interface that significantly reduce the binding energies.


Asunto(s)
ADN Primasa , Proteínas Hierro-Azufre , Oxidación-Reducción , Unión Proteica , ARN , ADN Primasa/metabolismo , ADN Primasa/química , ARN/metabolismo , ARN/química , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/metabolismo , ADN/metabolismo , ADN/química , Termodinámica , Modelos Moleculares
8.
J Am Chem Soc ; 146(14): 9583-9596, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38538061

RESUMEN

Primases are crucial enzymes for DNA replication, as they synthesize a short primer required for initiating DNA replication. We herein present time-resolved nuclear magnetic resonance (NMR) spectroscopy in solution and in the solid state to study the initial dinucleotide formation reaction of archaeal pRN1 primase. Our findings show that the helix-bundle domain (HBD) of pRN1 primase prepares the two substrates and then hands them over to the catalytic domain to initiate the reaction. By using nucleotide triphosphate analogues, the reaction is substantially slowed down, allowing us to study the initial dinucleotide formation in real time. We show that the sedimented protein-DNA complex remains active in the solid-state NMR rotor and that time-resolved 31P-detected cross-polarization experiments allow monitoring the kinetics of dinucleotide formation. The kinetics in the sedimented protein sample are comparable to those determined by solution-state NMR. Protein conformational changes during primer synthesis are observed in time-resolved 1H-detected experiments at fast magic-angle spinning frequencies (100 kHz). A significant number of spectral changes cluster in the HBD pointing to the importance of the HBD for positioning the nucleotides and the dinucleotide.


Asunto(s)
Carcinoma Papilar , Carcinoma de Células Renales , ADN Primasa , Replicación del ADN , Neoplasias de la Tiroides , ADN Primasa/química , Nucleótidos , Espectroscopía de Resonancia Magnética
9.
Crit Rev Biochem Mol Biol ; 56(1): 17-30, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33179522

RESUMEN

DNA replication forks are constantly challenged by DNA lesions induced by endogenous and exogenous sources. DNA damage tolerance mechanisms ensure that DNA replication continues with minimal effects on replication fork elongation either by using specialized DNA polymerases, which have the ability to replicate through the damaged template, or by skipping the damaged DNA, leaving it to be repaired after replication. These mechanisms are evolutionarily conserved in bacteria, yeast, and higher eukaryotes, and are paramount to ensure timely and faithful duplication of the genome. The Primase and DNA-directed Polymerase (PRIMPOL) is a recently discovered enzyme that possesses both primase and polymerase activities. PRIMPOL is emerging as a key player in DNA damage tolerance, particularly in vertebrate and human cells. Here, we review our current understanding of the function of PRIMPOL in DNA damage tolerance by focusing on the structural aspects that define its dual enzymatic activity, as well as on the mechanisms that control its chromatin recruitment and expression levels. We also focus on the latest findings on the mitochondrial and nuclear functions of PRIMPOL and on the impact of loss of these functions on genome stability and cell survival. Defining the function of PRIMPOL in DNA damage tolerance is becoming increasingly important in the context of human disease. In particular, we discuss recent evidence pointing at the PRIMPOL pathway as a novel molecular target to improve cancer cell response to DNA-damaging chemotherapy and as a predictive parameter to stratify patients in personalized cancer therapy.


Asunto(s)
Daño del ADN/genética , ADN Primasa/genética , ADN Primasa/metabolismo , Replicación del ADN/genética , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Enzimas Multifuncionales/genética , Enzimas Multifuncionales/metabolismo , Núcleo Celular/metabolismo , Supervivencia Celular/genética , Cromatina/metabolismo , ADN/genética , ADN/metabolismo , ADN Primasa/química , ADN Polimerasa Dirigida por ADN/química , Técnicas de Silenciamiento del Gen , Inestabilidad Genómica , Humanos , Mitocondrias/metabolismo , Enzimas Multifuncionales/química
10.
J Biol Chem ; 298(6): 101996, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35500649

RESUMEN

The T7 primase-helicase plays a pivotal role in the replication of T7 DNA. Using affinity isolation of peptide-nucleic acid crosslinks and mass spectrometry, we identify protein regions in the primase-helicase and T7 DNA polymerase that form contacts with the RNA primer and DNA template. The contacts between nucleic acids and the primase domain of the primase-helicase are centered in the RNA polymerase subdomain of the primase domain, in a cleft between the N-terminal subdomain and the topoisomerase-primase fold. We demonstrate that residues along a beta sheet in the N-terminal subdomain that contacts the RNA primer are essential for phage growth and primase activity in vitro. Surprisingly, we found mutations in the primase domain that had a dramatic effect on the helicase. Substitution of a residue conserved in other DnaG-like enzymes, R84A, abrogates both primase and helicase enzymatic activities of the T7 primase-helicase. Alterations in this residue also decrease binding of the primase-helicase to ssDNA. However, mass photometry measurements show that these mutations do not interfere with the ability of the protein to form the active hexamer.


Asunto(s)
Bacteriófago T7 , ADN Helicasas , ADN Primasa , ADN , Proteínas Virales , Secuencia de Aminoácidos , Bacteriófago T7/enzimología , ADN/metabolismo , ADN Helicasas/química , ADN Helicasas/metabolismo , ADN Primasa/química , ADN Primasa/genética , ADN Primasa/metabolismo , Mutación , Proteínas Virales/química , Proteínas Virales/metabolismo
11.
BMC Plant Biol ; 23(1): 467, 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37803262

RESUMEN

BACKGROUND: The mechanisms and regulation for DNA replication in plant organelles are largely unknown, as few proteins involved in replisome assembly have been biochemically studied. A primase-helicase dubbed Twinkle (T7 gp4-like protein with intramitochondrial nucleoid localization) unwinds double-stranded DNA in metazoan mitochondria and plant organelles. Twinkle in plants is a bifunctional enzyme with an active primase module. This contrast with animal Twinkle in which the primase module is inactive. The organellar primase-helicase of Arabidopsis thaliana (AtTwinkle) harbors a primase module (AtPrimase) that consists of an RNA polymerase domain (RPD) and a Zn + + finger domain (ZFD). RESULTS: Herein, we investigate the mechanisms by which AtTwinkle recognizes its templating sequence and how primer synthesis and coupling to the organellar DNA polymerases occurs. Biochemical data show that the ZFD of the AtPrimase module is responsible for template recognition, and this recognition is achieved by residues N163, R166, and K168. The role of the ZFD in template recognition was also corroborated by swapping the RPDs of bacteriophage T7 primase and AtPrimase with their respective ZFDs. A chimeric primase harboring the ZFD of T7 primase and the RPD of AtPrimase synthesizes ribonucleotides from the T7 primase recognition sequence and conversely, a chimeric primase harboring the ZFD of AtPrimase and the RPD of T7 primase synthesizes ribonucleotides from the AtPrimase recognition sequence. A chimera harboring the RPDs of bacteriophage T7 and the ZBD of AtTwinkle efficiently synthesizes primers for the plant organellar DNA polymerase. CONCLUSIONS: We conclude that the ZFD is responsible for recognizing a single-stranded sequence and for primer hand-off into the organellar DNA polymerases active site. The primase activity of plant Twinkle is consistent with phylogeny-based reconstructions that concluded that Twinkle´s last eukaryotic common ancestor (LECA) was an enzyme with primase and helicase activities. In plants, the primase domain is active, whereas the primase activity was lost in metazoans. Our data supports the notion that AtTwinkle synthesizes primers at the lagging-strand of the organellar replication fork.


Asunto(s)
Arabidopsis , ADN Primasa , Animales , ADN Primasa/genética , ADN Primasa/química , ADN Primasa/metabolismo , ADN Helicasas/química , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Arabidopsis/metabolismo , Mitocondrias/metabolismo , Dedos de Zinc , Ribonucleótidos , Replicación del ADN , Bacteriófago T7/genética
12.
Nucleic Acids Res ; 49(20): 11447-11458, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34718733

RESUMEN

DNA-protein interactions play essential roles in all living cells. Understanding of how features embedded in the DNA sequence affect specific interactions with proteins is both challenging and important, since it may contribute to finding the means to regulate metabolic pathways involving DNA-protein interactions. Using a massive experimental benchmark dataset of binding scores for DNA sequences and a machine learning workflow, we describe the binding to DNA of T7 primase, as a model system for specific DNA-protein interactions. Effective binding of T7 primase to its specific DNA recognition sequences triggers the formation of RNA primers that serve as Okazaki fragment start sites during DNA replication.


Asunto(s)
ADN Primasa/química , ADN/química , Motivos de Nucleótidos , Sitios de Unión , ADN/metabolismo , ADN Primasa/metabolismo , Aprendizaje Automático , Unión Proteica
13.
Nucleic Acids Res ; 49(4): 2179-2191, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33533925

RESUMEN

Replication forks often stall at damaged DNA. To overcome these obstructions and complete the DNA duplication in a timely fashion, replication can be restarted downstream of the DNA lesion. In mammalian cells, this repriming of replication can be achieved through the activities of primase and polymerase PrimPol. PrimPol is stimulated in DNA synthesis through interaction with PolDIP2, however the exact mechanism of this PolDIP2-dependent stimulation is still unclear. Here, we show that PrimPol uses a flexible loop to interact with the C-terminal ApaG-like domain of PolDIP2, and that this contact is essential for PrimPol's enhanced processivity. PolDIP2 increases primer-template and dNTP binding affinities of PrimPol, which concomitantly enhances its nucleotide incorporation efficiency. This stimulation is dependent on a unique arginine cluster in PolDIP2. Since the polymerase activity of PrimPol alone is very limited, this mechanism, where the affinity for dNTPs gets increased by PolDIP2 binding, might be critical for the in vivo function of PrimPol in tolerating DNA lesions at physiological nucleotide concentrations.


Asunto(s)
Arginina/química , ADN Primasa/química , ADN Polimerasa Dirigida por ADN/química , ADN/biosíntesis , Enzimas Multifuncionales/química , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Secuencias de Aminoácidos , ADN Primasa/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Desoxirribonucleótidos/química , Desoxirribonucleótidos/metabolismo , Modelos Moleculares , Enzimas Multifuncionales/metabolismo , Unión Proteica
14.
Nucleic Acids Res ; 49(8): 4599-4612, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33849056

RESUMEN

The eukaryotic replisome is comprised of three family-B DNA polymerases (Polα, δ and ϵ). Polα forms a stable complex with primase to synthesize short RNA-DNA primers, which are subsequently elongated by Polδ and Polϵ in concert with proliferating cell nuclear antigen (PCNA). In some species of archaea, family-D DNA polymerase (PolD) is the only DNA polymerase essential for cell viability, raising the question of how it alone conducts the bulk of DNA synthesis. We used a hyperthermophilic archaeon, Thermococcus kodakarensis, to demonstrate that PolD connects primase to the archaeal replisome before interacting with PCNA. Whereas PolD stably connects primase to GINS, a component of CMG helicase, cryo-EM analysis indicated a highly flexible PolD-primase complex. A conserved hydrophobic motif at the C-terminus of the DP2 subunit of PolD, a PIP (PCNA-Interacting Peptide) motif, was critical for the interaction with primase. The dissociation of primase was induced by DNA-dependent binding of PCNA to PolD. Point mutations in the alternative PIP-motif of DP2 abrogated the molecular switching that converts the archaeal replicase from de novo to processive synthesis mode.


Asunto(s)
Proteínas Arqueales/metabolismo , ADN Helicasas/metabolismo , ADN Polimerasa III/metabolismo , ADN Primasa/química , Antígeno Nuclear de Célula en Proliferación/metabolismo , Thermococcus/metabolismo , Secuencias de Aminoácidos , Proteínas Arqueales/química , Cromatografía en Gel , ADN Helicasas/genética , ADN Polimerasa III/química , ADN Primasa/genética , ADN Primasa/metabolismo , Escherichia coli/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Mutagénesis Sitio-Dirigida , Electroforesis en Gel de Poliacrilamida Nativa , Antígeno Nuclear de Célula en Proliferación/genética , Unión Proteica , Proteínas Recombinantes , Resonancia por Plasmón de Superficie , Thermococcus/genética
15.
Nucleic Acids Res ; 48(6): 3156-3164, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32009150

RESUMEN

The hexametric T7 helicase (gp4) adopts a spiral lock-washer form and encircles a coil-like DNA (tracking) strand with two nucleotides bound to each subunit. However, the chemo-mechanical coupling mechanism in unwinding has yet to be elucidated. Here, we utilized nanotensioner-enhanced Förster resonance energy transfer with one nucleotide precision to investigate gp4-induced unwinding of DNA that contains an abasic lesion. We observed that the DNA unwinding activity of gp4 is hindered but not completely blocked by abasic lesions. Gp4 moves back and forth repeatedly when it encounters an abasic lesion, whereas it steps back only occasionally when it unwinds normal DNA. We further observed that gp4 translocates on the tracking strand in step sizes of one to four nucleotides. We propose that a hypothetical intermediate conformation of the gp4-DNA complex during DNA unwinding can help explain how gp4 molecules pass lesions, providing insights into the unwinding dynamics of gp4.


Asunto(s)
Bacteriófago T7/genética , ADN Helicasas/genética , ADN Primasa/genética , ADN/genética , Bacteriófago T7/química , ADN/química , ADN Primasa/química , Transferencia Resonante de Energía de Fluorescencia , Cinética , Conformación Molecular , Nucleótidos/química , Nucleótidos/genética
16.
J Biol Chem ; 295(50): 17298-17309, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33055234

RESUMEN

The faithful segregation, or "partition," of many low-copy number bacterial plasmids is driven by plasmid-encoded ATPases that are represented by the P1 plasmid ParA protein. ParA binds to the bacterial nucleoid via an ATP-dependent nonspecific DNA (nsDNA)-binding activity, which is essential for partition. ParA also has a site-specific DNA-binding activity to the par operator (parOP), which requires either ATP or ADP, and which is essential for it to act as a transcriptional repressor but is dispensable for partition. Here we examine how DNA binding by ParA contributes to the relative distribution of its plasmid partition and repressor activities, using a ParA with an alanine substitution at Arg351, a residue previously predicted to participate in site-specific DNA binding. In vivo, the parAR351A allele is compromised for partition, but its repressor activity is dramatically improved so that it behaves as a "super-repressor." In vitro, ParAR351A binds and hydrolyzes ATP, and undergoes a specific conformational change required for nsDNA binding, but its nsDNA-binding activity is significantly damaged. This defect in turn significantly reduces the assembly and stability of partition complexes formed by the interaction of ParA with ParB, the centromere-binding protein, and DNA. In contrast, the R351A change shows only a mild defect in site-specific DNA binding. We conclude that the partition defect is due to altered nsDNA binding kinetics and affinity for the bacterial chromosome. Furthermore, the super-repressor phenotype is explained by an increased pool of non-nucleoid bound ParA that is competent to bind parOP and repress transcription.


Asunto(s)
Bacteriófago P1/metabolismo , Cromosomas Bacterianos/metabolismo , ADN Bacteriano/metabolismo , Escherichia coli/metabolismo , Proteínas Virales/metabolismo , Sustitución de Aminoácidos , Bacteriófago P1/química , Bacteriófago P1/genética , Cromosomas Bacterianos/química , Cromosomas Bacterianos/genética , ADN Primasa/química , ADN Primasa/genética , ADN Primasa/metabolismo , ADN Bacteriano/química , ADN Bacteriano/genética , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/virología , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Mutación Missense , Regiones Operadoras Genéticas , Proteínas Virales/química , Proteínas Virales/genética
17.
Phys Rev Lett ; 127(13): 138101, 2021 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-34623846

RESUMEN

The spatiotemporal organization of bacterial cells is crucial for the active segregation of replicating chromosomes. In several species, including Caulobacter crescentus, the ATPase ParA binds to DNA and forms a gradient along the long cell axis. The ParB partition complex on the newly replicated chromosome translocates up this ParA gradient, thereby contributing to chromosome segregation. A DNA-relay mechanism-deriving from the elasticity of the fluctuating chromosome-has been proposed as the driving force for this cargo translocation, but a mechanistic theoretical description remains elusive. Here, we propose a minimal model to describe force generation by the DNA-relay mechanism over a broad range of operational conditions. Conceptually, we identify four distinct force-generation regimes characterized by their dependence on chromosome fluctuations. These relay force regimes arise from an interplay of the imposed ParA gradient, chromosome fluctuations, and an emergent friction force due to chromosome-cargo interactions.


Asunto(s)
ADN Bacteriano/metabolismo , Modelos Biológicos , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Transporte Biológico , Caulobacter crescentus/genética , Caulobacter crescentus/metabolismo , Segregación Cromosómica , Cromosomas Bacterianos , ADN Primasa/química , ADN Primasa/genética , ADN Primasa/metabolismo , ADN Bacteriano/química , ADN Bacteriano/genética
18.
Mol Cell ; 52(4): 566-73, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24267451

RESUMEN

DNA damage can stall the DNA replication machinery, leading to genomic instability. Thus, numerous mechanisms exist to complete genome duplication in the absence of a pristine DNA template, but identification of the enzymes involved remains incomplete. Here, we establish that Primase-Polymerase (PrimPol; CCDC111), an archaeal-eukaryotic primase (AEP) in eukaryotic cells, is involved in chromosomal DNA replication. PrimPol is required for replication fork progression on ultraviolet (UV) light-damaged DNA templates, possibly mediated by its ability to catalyze translesion synthesis (TLS) of these lesions. This PrimPol UV lesion bypass pathway is not epistatic with the Pol η-dependent pathway and, as a consequence, protects xeroderma pigmentosum variant (XP-V) patient cells from UV-induced cytotoxicity. In addition, we establish that PrimPol is also required for efficient replication fork progression during an unperturbed S phase. These and other findings indicate that PrimPol is an important player in replication fork progression in eukaryotic cells.


Asunto(s)
Cromosomas Humanos/genética , Aductos de ADN/genética , ADN Primasa/fisiología , Replicación del ADN , ADN Polimerasa Dirigida por ADN/fisiología , Enzimas Multifuncionales/fisiología , Secuencia de Aminoácidos , Animales , Proliferación Celular , Supervivencia Celular , Pollos , Aductos de ADN/química , Aductos de ADN/metabolismo , Daño del ADN , ADN Primasa/química , ADN de Cadena Simple/química , ADN Polimerasa Dirigida por ADN/química , Puntos de Control de la Fase G2 del Ciclo Celular , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Enzimas Multifuncionales/química , Rayos Ultravioleta , Xenopus
19.
Mol Cell ; 52(4): 541-53, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24207056

RESUMEN

We describe a second primase in human cells, PrimPol, which has the ability to start DNA chains with deoxynucleotides unlike regular primases, which use exclusively ribonucleotides. Moreover, PrimPol is also a DNA polymerase tailored to bypass the most common oxidative lesions in DNA, such as abasic sites and 8-oxoguanine. Subcellular fractionation and immunodetection studies indicated that PrimPol is present in both nuclear and mitochondrial DNA compartments. PrimPol activity is detectable in mitochondrial lysates from human and mouse cells but is absent from mitochondria derived from PRIMPOL knockout mice. PRIMPOL gene silencing or ablation in human and mouse cells impaired mitochondrial DNA replication. On the basis of the synergy observed with replicative DNA polymerases Polγ and Polε, PrimPol is proposed to facilitate replication fork progression by acting as a translesion DNA polymerase or as a specific DNA primase reinitiating downstream of lesions that block synthesis during both mitochondrial and nuclear DNA replication.


Asunto(s)
ADN Primasa/fisiología , Replicación del ADN , ADN Polimerasa Dirigida por ADN/fisiología , Enzimas Multifuncionales/fisiología , Secuencia de Aminoácidos , Animales , Ácido Apurínico/química , Secuencia de Bases , Dominio Catalítico , Núcleo Celular/enzimología , ADN Polimerasa II/química , ADN Polimerasa gamma , ADN Primasa/química , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , ADN Polimerasa Dirigida por ADN/química , Desoxiadenosinas/química , Desoxirribonucleótidos/química , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Noqueados , Mitocondrias/enzimología , Datos de Secuencia Molecular , Enzimas Multifuncionales/química
20.
Nucleic Acids Res ; 47(7): 3422-3433, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30759238

RESUMEN

The developmental asymmetry of fission yeast daughter cells derives from inheriting 'older Watson' versus 'older Crick' DNA strand from the parental cell, strands that are complementary but not identical with each other. A novel DNA strand-specific 'imprint', installed during DNA replication at the mating-type locus (mat1), imparts competence for cell type inter-conversion to one of the two chromosome replicas. The catalytic subunit of DNA Polymerase α (Polα) has been implicated in the imprinting process. Based on its known biochemical function, Polα might install the mat1 imprint during lagging strand synthesis. The nature of the imprint is not clear: it is either a nick or a ribonucleotide insertion. Our investigations do not support a direct role of Polα in nicking through putative endonuclease domains but confirm its indirect role in installing an alkali-labile moiety as the imprint. While ruling out the role of the primase subunit of Polα holoenzyme, we find that mutations in the Polα-recruitment and putative primase homology domain in Mcm10/Cdc23 abrogate the ribonucleotide imprint formation. These results, while confirming the ribonucleotide nature of the imprint suggest the possibility of a direct role of Mcm10/Cdc23 in installing it in cooperation with Polα and Swi1.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , ADN Polimerasa I/metabolismo , Replicación del ADN/genética , Genes del Tipo Sexual de los Hongos/genética , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Ribonucleótidos/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Dominio Catalítico , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , ADN Polimerasa I/química , ADN Polimerasa I/genética , ADN Primasa/química , ADN Primasa/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Mantenimiento de Minicromosoma/química , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA