Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FEBS J ; 291(8): 1813-1829, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38335062

RESUMO

Eukaryotic DNA replication depends on the primosome - a complex of DNA polymerase alpha (Pol α) and primase - to initiate DNA synthesis by polymerisation of an RNA-DNA primer. Primer synthesis requires the tight coordination of primase and polymerase activities. Recent cryo-electron microscopy (cryoEM) analyses have elucidated the extensive conformational transitions required for RNA primer handover between primase and Pol α and primer elongation by Pol α. Because of the intrinsic flexibility of the primosome, however, structural information about the initiation of RNA primer synthesis is still lacking. Here, we capture cryoEM snapshots of the priming reaction to reveal the conformational trajectory of the human primosome that brings DNA primase subunits 1 and 2 (PRIM1 and PRIM2, respectively) together, poised for RNA synthesis. Furthermore, we provide experimental evidence for the continuous association of primase subunit PRIM2 with the RNA primer during primer synthesis, and for how both initiation and termination of RNA primer polymerisation are licenced by specific rearrangements of DNA polymerase alpha catalytic subunit (POLA1), the polymerase subunit of Pol α. Our findings fill a critical gap in our understanding of the conformational changes that underpin the synthesis of the RNA primer by the primosome. Together with existing evidence, they provide a complete description of the structural dynamics of the human primosome during DNA replication initiation.


Assuntos
DNA Polimerase I , DNA Primase , Humanos , DNA Primase/genética , DNA Primase/metabolismo , Microscopia Crioeletrônica , DNA Polimerase I/genética , RNA , Replicação do DNA
2.
Protein Sci ; 31(2): 333-344, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34719824

RESUMO

The molecular mechanisms that drive the infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-the causative agent of coronavirus disease 2019 (COVID-19)-are under intense current scrutiny to understand how the virus operates and to uncover ways in which the disease can be prevented or alleviated. Recent proteomic screens of the interactions between viral and host proteins have identified the human proteins targeted by SARS-CoV-2. The DNA polymerase α (Pol α)-primase complex or primosome-responsible for initiating DNA synthesis during genomic duplication-was identified as a target of nonstructural protein 1 (nsp1), a major virulence factor in the SARS-CoV-2 infection. Here, we validate the published reports of the interaction of nsp1 with the primosome by demonstrating direct binding with purified recombinant components and providing a biochemical characterization of their interaction. Furthermore, we provide a structural basis for the interaction by elucidating the cryo-electron microscopy structure of nsp1 bound to the primosome. Our findings provide biochemical evidence for the reported targeting of Pol α by the virulence factor nsp1 and suggest that SARS-CoV-2 interferes with Pol α's putative role in the immune response during the viral infection.


Assuntos
COVID-19 , SARS-CoV-2 , Proteínas não Estruturais Virais , Microscopia Crioeletrônica , DNA Polimerase I , DNA Primase , Humanos , Proteômica , Proteínas não Estruturais Virais/genética , Fatores de Virulência
3.
Science ; 369(6504)2020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32764038

RESUMO

Sulfolobus acidocaldarius is the closest experimentally tractable archaeal relative of eukaryotes and, despite lacking obvious cyclin-dependent kinase and cyclin homologs, has an ordered eukaryote-like cell cycle with distinct phases of DNA replication and division. Here, in exploring the mechanism of cell division in S. acidocaldarius, we identify a role for the archaeal proteasome in regulating the transition from the end of one cell cycle to the beginning of the next. Further, we identify the archaeal ESCRT-III homolog, CdvB, as a key target of the proteasome and show that its degradation triggers division by allowing constriction of the CdvB1:CdvB2 ESCRT-III division ring. These findings offer a minimal mechanism for ESCRT-III-mediated membrane remodeling and point to a conserved role for the proteasome in eukaryotic and archaeal cell cycle control.


Assuntos
Proteínas Arqueais/fisiologia , Divisão Celular , Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Sulfolobus acidocaldarius/citologia , Proteínas Arqueais/química , Bortezomib/química , Bortezomib/farmacologia , Complexos Endossomais de Distribuição Requeridos para Transporte/química , Modelos Moleculares , Complexo de Endopeptidases do Proteassoma/química , Inibidores de Proteassoma/química , Inibidores de Proteassoma/farmacologia , Proteólise , Sulfolobus acidocaldarius/efeitos dos fármacos , Sulfolobus acidocaldarius/enzimologia
4.
EMBO J ; 39(18): e104185, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32705708

RESUMO

Regions of the genome with the potential to form secondary DNA structures pose a frequent and significant impediment to DNA replication and must be actively managed in order to preserve genetic and epigenetic integrity. How the replisome detects and responds to secondary structures is poorly understood. Here, we show that a core component of the fork protection complex in the eukaryotic replisome, Timeless, harbours in its C-terminal region a previously unappreciated DNA-binding domain that exhibits specific binding to G-quadruplex (G4) DNA structures. We show that this domain contributes to maintaining processive replication through G4-forming sequences, and exhibits partial redundancy with an adjacent PARP-binding domain. Further, this function of Timeless requires interaction with and activity of the helicase DDX11. Loss of both Timeless and DDX11 causes epigenetic instability at G4-forming sequences and DNA damage. Our findings indicate that Timeless contributes to the ability of the replisome to sense replication-hindering G4 formation and ensures the prompt resolution of these structures by DDX11 to maintain processive DNA synthesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , RNA Helicases DEAD-box/metabolismo , Dano ao DNA , DNA Helicases/metabolismo , Replicação do DNA , Quadruplex G , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular , RNA Helicases DEAD-box/genética , DNA Helicases/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Domínios Proteicos
6.
ACS Chem Biol ; 14(9): 1904-1912, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31479243

RESUMO

Nucleoside analogues are widely used in clinical practice as chemotherapy drugs. Arabinose nucleoside derivatives such as fludarabine are effective in the treatment of patients with acute and chronic leukemias and non-Hodgkin's lymphomas. Although nucleoside analogues are generally known to function by inhibiting DNA synthesis in rapidly proliferating cells, the identity of their in vivo targets and mechanism of action are often not known in molecular detail. Here we provide a structural basis for arabinose nucleotide-mediated inhibition of human primase, the DNA-dependent RNA polymerase responsible for initiation of DNA synthesis in DNA replication. Our data suggest ways in which the chemical structure of fludarabine could be modified to improve its specificity and affinity toward primase, possibly leading to less toxic and more effective therapeutic agents.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Antineoplásicos/química , Antivirais/química , DNA Primase/antagonistas & inibidores , Vidarabina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Antineoplásicos/metabolismo , Antivirais/metabolismo , Domínio Catalítico , Cristalografia por Raios X , DNA Primase/química , DNA Primase/metabolismo , Ensaios Enzimáticos , Humanos , Ligação Proteica , Vidarabina/química , Vidarabina/metabolismo
7.
Open Biol ; 7(11)2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29167311

RESUMO

A dynamic multi-protein assembly known as the replisome is responsible for DNA synthesis in eukaryotic cells. In yeast, the hub protein Ctf4 bridges DNA helicase and DNA polymerase and recruits factors with roles in metabolic processes coupled to DNA replication. An important question in DNA replication is the extent to which the molecular architecture of the replisome is conserved between yeast and higher eukaryotes. Here, we describe the biochemical basis for the interaction of the human CTF4-orthologue AND-1 with DNA polymerase α (Pol α)/primase, the replicative polymerase that initiates DNA synthesis. AND-1 has maintained the trimeric structure of yeast Ctf4, driven by its conserved SepB domain. However, the primary interaction of AND-1 with Pol α/primase is mediated by its C-terminal HMG box, unique to mammalian AND-1, which binds the B subunit, at the same site targeted by the SV40 T-antigen for viral replication. In addition, we report a novel DNA-binding activity in AND-1, which might promote the correct positioning of Pol α/primase on the lagging-strand template at the replication fork. Our findings provide a biochemical basis for the specific interaction between two critical components of the human replisome, and indicate that important principles of replisome architecture have changed significantly in evolution.


Assuntos
DNA Polimerase I/metabolismo , DNA Primase/metabolismo , Proteínas de Ligação a DNA/metabolismo , Domínios HMG-Box , Sítios de Ligação , Biologia Computacional , Humanos , Modelos Moleculares , Ligação Proteica
8.
Nat Commun ; 8(1): 1718, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-29167441

RESUMO

DNA replication depends on primase, the specialised polymerase responsible for synthesis of the RNA primers that are elongated by the replicative DNA polymerases. In eukaryotic and archaeal replication, primase is a heterodimer of two subunits, PriS and PriL. Recently, a third primase subunit named PriX was identified in the archaeon Sulfolobus solfataricus. PriX is essential for primer synthesis and is structurally related to the Fe-S cluster domain of eukaryotic PriL. Here we show that PriX contains a nucleotide-binding site required for primer synthesis, and demonstrate equivalence of nucleotide-binding residues in PriX with eukaryotic PriL residues that are known to be important for primer synthesis. A primase chimera, where PriX is fused to a truncated version of PriL lacking the Fe-S cluster domain retains wild-type levels of primer synthesis. Our evidence shows that PriX has replaced PriL as the subunit that endows primase with the unique ability to initiate nucleic acid synthesis. Importantly, our findings reveal that the Fe-S cluster is not required for primer synthesis.

9.
Mol Cell ; 67(5): 867-881.e7, 2017 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-28757209

RESUMO

Brca2 deficiency causes Mre11-dependent degradation of nascent DNA at stalled forks, leading to cell lethality. To understand the molecular mechanisms underlying this process, we isolated Xenopus laevis Brca2. We demonstrated that Brca2 protein prevents single-stranded DNA gap accumulation at replication fork junctions and behind them by promoting Rad51 binding to replicating DNA. Without Brca2, forks with persistent gaps are converted by Smarcal1 into reversed forks, triggering extensive Mre11-dependent nascent DNA degradation. Stable Rad51 nucleofilaments, but not RPA or Rad51T131P mutant proteins, directly prevent Mre11-dependent DNA degradation. Mre11 inhibition instead promotes reversed fork accumulation in the absence of Brca2. Rad51 directly interacts with the Pol α N-terminal domain, promoting Pol α and δ binding to stalled replication forks. This interaction likely promotes replication fork restart and gap avoidance. These results indicate that Brca2 and Rad51 prevent formation of abnormal DNA replication intermediates, whose processing by Smarcal1 and Mre11 predisposes to genome instability.


Assuntos
Proteína BRCA2/metabolismo , Replicação do DNA , DNA/biossíntese , Rad51 Recombinase/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animais , Proteína BRCA2/genética , Sítios de Ligação , DNA/genética , DNA Helicases/genética , DNA Helicases/metabolismo , DNA Polimerase I/metabolismo , DNA Polimerase III/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/metabolismo , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Feminino , Instabilidade Genômica , Humanos , Proteína Homóloga a MRE11 , Masculino , Mutação , Ligação Proteica , Rad51 Recombinase/genética , Origem de Replicação , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Tempo , Proteínas de Xenopus/genética , Xenopus laevis/genética
10.
Nucleic Acids Res ; 45(9): 5555-5563, 2017 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-28334766

RESUMO

Human Timeless is involved in replication fork stabilization, S-phase checkpoint activation and establishment of sister chromatid cohesion. In the cell, Timeless forms a constitutive heterodimeric complex with Tipin. Here we present the 1.85 Å crystal structure of a large N-terminal segment of human Timeless, spanning amino acids 1-463, and we show that this region of human Timeless harbours a partial binding site for Tipin. Furthermore, we identify minimal regions of the two proteins that are required for the formation of a stable Timeless-Tipin complex and provide evidence that the Timeless-Tipin interaction is based on a composite binding interface comprising different domains of Timeless.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Fenômenos Biofísicos , Reagentes de Ligações Cruzadas/metabolismo , Cristalografia por Raios X , Proteínas de Ligação a DNA , Humanos , Espectrometria de Massas , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Multimerização Proteica , Homologia Estrutural de Proteína
11.
Mol Cell ; 63(3): 385-96, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27397685

RESUMO

Replisome assembly at eukaryotic replication forks connects the DNA helicase to DNA polymerases and many other factors. The helicase binds the leading-strand polymerase directly, but is connected to the Pol α lagging-strand polymerase by the trimeric adaptor Ctf4. Here, we identify new Ctf4 partners in addition to Pol α and helicase, all of which contain a "Ctf4-interacting-peptide" or CIP-box. Crystallographic analysis classifies CIP-boxes into two related groups that target different sites on Ctf4. Mutations in the CIP-box motifs of the Dna2 nuclease or the rDNA-associated protein Tof2 do not perturb DNA synthesis genome-wide, but instead lead to a dramatic shortening of chromosome 12 that contains the large array of rDNA repeats. Our data reveal unexpected complexity of Ctf4 function, as a hub that connects multiple accessory factors to the replisome. Most strikingly, Ctf4-dependent recruitment of CIP-box proteins couples other processes to DNA synthesis, including rDNA copy-number regulation.


Assuntos
Cromossomos Fúngicos/enzimologia , DNA Helicases/metabolismo , DNA Fúngico/biossíntese , DNA Ribossômico/biossíntese , Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fase S , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Sítios de Ligação , Cromossomos Fúngicos/genética , DNA Helicases/genética , DNA Polimerase I/metabolismo , DNA Fúngico/genética , DNA Ribossômico/genética , Proteínas de Ligação a DNA/genética , Dosagem de Genes , Peptídeos e Proteínas de Sinalização Intracelular/genética , Modelos Moleculares , Complexos Multiproteicos , Mutação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/genética , Relação Estrutura-Atividade
12.
Nat Commun ; 6: 8163, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-26348592

RESUMO

In eukaryotes, the covalent attachment of ubiquitin chains directs substrates to the proteasome for degradation. Recently, ubiquitin-like modifications have also been described in the archaeal domain of life. It has subsequently been hypothesized that ubiquitin-like proteasomal degradation might also operate in these microbes, since all archaeal species utilize homologues of the eukaryotic proteasome. Here we perform a structural and biochemical analysis of a ubiquitin-like modification pathway in the archaeon Sulfolobus acidocaldarius. We reveal that this modifier is homologous to the eukaryotic ubiquitin-related modifier Urm1, considered to be a close evolutionary relative of the progenitor of all ubiquitin-like proteins. Furthermore we demonstrate that urmylated substrates are recognized and processed by the archaeal proteasome, by virtue of a direct interaction with the modifier. Thus, the regulation of protein stability by Urm1 and the proteasome in archaea is likely representative of an ancient pathway from which eukaryotic ubiquitin-mediated proteolysis has evolved.


Assuntos
Proteínas Arqueais/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Sulfolobus acidocaldarius/genética , Ubiquitinas/genética , Proteínas Arqueais/metabolismo , Cromatografia em Gel , Cromatografia Líquida , Dicroísmo Circular , Cristalografia por Raios X , Espectrometria de Massas , Microscopia Eletrônica , Complexo de Endopeptidases do Proteassoma/ultraestrutura , Proteólise , Sulfolobus acidocaldarius/metabolismo , Ubiquitinas/metabolismo
13.
Nature ; 510(7504): 293-297, 2014 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-24805245

RESUMO

Efficient duplication of the genome requires the concerted action of helicase and DNA polymerases at replication forks to avoid stalling of the replication machinery and consequent genomic instability. In eukaryotes, the physical coupling between helicase and DNA polymerases remains poorly understood. Here we define the molecular mechanism by which the yeast Ctf4 protein links the Cdc45-MCM-GINS (CMG) DNA helicase to DNA polymerase α (Pol α) within the replisome. We use X-ray crystallography and electron microscopy to show that Ctf4 self-associates in a constitutive disk-shaped trimer. Trimerization depends on a ß-propeller domain in the carboxy-terminal half of the protein, which is fused to a helical extension that protrudes from one face of the trimeric disk. Critically, Pol α and the CMG helicase share a common mechanism of interaction with Ctf4. We show that the amino-terminal tails of the catalytic subunit of Pol α and the Sld5 subunit of GINS contain a conserved Ctf4-binding motif that docks onto the exposed helical extension of a Ctf4 protomer within the trimer. Accordingly, we demonstrate that one Ctf4 trimer can support binding of up to three partner proteins, including the simultaneous association with both Pol α and GINS. Our findings indicate that Ctf4 can couple two molecules of Pol α to one CMG helicase within the replisome, providing a new model for lagging-strand synthesis in eukaryotes that resembles the emerging model for the simpler replisome of Escherichia coli. The ability of Ctf4 to act as a platform for multivalent interactions illustrates a mechanism for the concurrent recruitment of factors that act together at the fork.


Assuntos
DNA Helicases/metabolismo , DNA Polimerase I/metabolismo , Replicação do DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/metabolismo , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Multimerização Proteica , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Domínio Catalítico , Sequência Conservada , Cristalografia por Raios X , DNA Helicases/química , DNA Helicases/ultraestrutura , DNA Polimerase I/química , DNA Polimerase I/ultraestrutura , Proteínas de Ligação a DNA/ultraestrutura , Microscopia Eletrônica , Proteínas de Manutenção de Minicromossomo/química , Proteínas de Manutenção de Minicromossomo/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/ultraestrutura
14.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 4): 1166-72, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24699660

RESUMO

Aspartate α-decarboxylase is a pyruvoyl-dependent decarboxylase required for the production of ß-alanine in the bacterial pantothenate (vitamin B5) biosynthesis pathway. The pyruvoyl group is formed via the intramolecular rearrangement of a serine residue to generate a backbone ester intermediate which is cleaved to generate an N-terminal pyruvoyl group. Site-directed mutagenesis of residues adjacent to the active site, including Tyr22, Thr57 and Tyr58, reveals that only mutation of Thr57 leads to changes in the degree of post-translational activation. The crystal structure of the site-directed mutant T57V is consistent with a non-rearranged backbone, supporting the hypothesis that Thr57 is required for the formation of the ester intermediate in activation.


Assuntos
Escherichia coli/enzimologia , Glutamato Descarboxilase/química , Ativação Enzimática , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Modelos Moleculares , Mutação , Estrutura Terciária de Proteína , Treonina/genética , Treonina/metabolismo
15.
Proc Natl Acad Sci U S A ; 110(40): 15961-6, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043831

RESUMO

Initiation of DNA synthesis in genomic duplication depends on primase, the DNA-dependent RNA polymerase that synthesizes de novo the oligonucleotides that prime DNA replication. Due to the discontinuous nature of DNA replication, primase activity on the lagging strand is required throughout the replication process. In eukaryotic cells, the presence of primase at the replication fork is secured by its physical association with DNA polymerase α (Pol α), which extends the RNA primer with deoxynucleotides. Our knowledge of the mechanism that primes DNA synthesis is very limited, as structural information for the eukaryotic enzyme has proved difficult to obtain. Here, we describe the crystal structure of human primase in heterodimeric form consisting of full-length catalytic subunit and a C-terminally truncated large subunit. We exploit the crystallographic model to define the architecture of its nucleotide elongation site and to show that the small subunit integrates primer initiation and elongation within the same set of functional residues. Furthermore, we define in atomic detail the mode of association of primase to Pol α, the critical interaction that keeps primase tethered to the eukaryotic replisome.


Assuntos
DNA Polimerase I/química , DNA Primase/química , DNA Primase/metabolismo , Replicação do DNA/fisiologia , Modelos Moleculares , Conformação Proteica , Clonagem Molecular , Cristalização , Cristalografia por Raios X , DNA Polimerase I/metabolismo , Dimerização , Polarização de Fluorescência , Humanos
16.
Elife ; 2: e00482, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23599895

RESUMO

The DNA Polymerase α (Pol α)/primase complex initiates DNA synthesis in eukaryotic replication. In the complex, Pol α and primase cooperate in the production of RNA-DNA oligonucleotides that prime synthesis of new DNA. Here we report crystal structures of the catalytic core of yeast Pol α in unliganded form, bound to an RNA primer/DNA template and extending an RNA primer with deoxynucleotides. We combine the structural analysis with biochemical and computational data to demonstrate that Pol α specifically recognizes the A-form RNA/DNA helix and that the ensuing synthesis of B-form DNA terminates primer synthesis. The spontaneous release of the completed RNA-DNA primer by the Pol α/primase complex simplifies current models of primer transfer to leading- and lagging strand polymerases. The proposed mechanism of nucleotide polymerization by Pol α might contribute to genomic stability by limiting the amount of inaccurate DNA to be corrected at the start of each Okazaki fragment. DOI:http://dx.doi.org/10.7554/eLife.00482.001.


Assuntos
DNA Polimerase I/metabolismo , DNA Fúngico/biossíntese , Saccharomyces cerevisiae/genética , Domínio Catalítico , DNA Fúngico/química , Modelos Moleculares , Conformação de Ácido Nucleico , Saccharomyces cerevisiae/enzimologia
17.
J Biol Chem ; 287(28): 23740-7, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22593576

RESUMO

The DNA polymerase α-primase complex forms an essential part of the eukaryotic replisome. The catalytic subunits of primase and pol α synthesize composite RNA-DNA primers that initiate the leading and lagging DNA strands at replication forks. The physical basis and physiological significance of tethering primase to the eukaryotic replisome via pol α remain poorly characterized. We have identified a short conserved motif at the extreme C terminus of pol α that is critical for interaction of the yeast ortholog pol1 with primase. We show that truncation of the C-terminal residues 1452-1468 of Pol1 abrogates the interaction with the primase, as does mutation to alanine of the invariant amino acid Phe(1463). Conversely, a pol1 peptide spanning the last 16 residues binds primase with high affinity, and the equivalent peptide from human Pol α binds primase in an analogous fashion. These in vitro data are mirrored by experiments in yeast cells, as primase does not interact in cell extracts with pol1 that either terminates at residue 1452 or has the F1463A mutation. The ability to disrupt the association between primase and pol α allowed us to assess the physiological significance of primase being tethered to the eukaryotic replisome in this way. We find that the F1463A mutation in Pol1 renders yeast cells dependent on the S phase checkpoint, whereas truncation of Pol1 at amino acid 1452 blocks yeast cell proliferation. These findings indicate that tethering of primase to the replisome by pol α is critical for the normal action of DNA replication forks in eukaryotic cells.


Assuntos
DNA Polimerase I/metabolismo , DNA Primase/metabolismo , Replicação do DNA/genética , Células Eucarióticas/metabolismo , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Sequência Conservada/genética , DNA Polimerase I/química , DNA Polimerase I/genética , DNA Primase/química , DNA Primase/genética , Humanos , Immunoblotting , Imunoprecipitação , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos
18.
Artigo em Inglês | MEDLINE | ID: mdl-22505409

RESUMO

The crystal structure of the Asn72Ala site-directed mutant of Escherichia coli aspartate α-decarboxylase (ADC) has been determined at 1.7 Å resolution. The refined structure is consistent with the presence of a hydrolysis product serine in the active site in place of the pyruvoyl group required for catalysis, which suggests that the role of Asn72 is to protect the ester formed during ADC activation from hydrolysis. In previously determined structures of activated ADC, including the wild type and other site-directed mutants, the C-terminal region of the protein is disordered, but in the Asn72Ala mutant these residues are ordered owing to an interaction with the active site of the neighbouring symmetry-related multimer.


Assuntos
Escherichia coli/enzimologia , Glutamato Descarboxilase/química , Ativação Enzimática , Glutamato Descarboxilase/genética , Modelos Moleculares , Mutação , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Especificidade por Substrato
19.
Nucleic Acids Res ; 39(18): 8187-99, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21715379

RESUMO

The Pol α/primase complex or primosome is the primase/polymerase complex that initiates nucleic acid synthesis during eukaryotic replication. Within the primosome, the primase synthesizes short RNA primers that undergo limited extension by Pol α. The resulting RNA-DNA primers are utilized by Pol δ and Pol ε for processive elongation on the lagging and leading strands, respectively. Despite its importance, the mechanism of RNA-DNA primer synthesis remains poorly understood. Here, we describe a structural model of the yeast primosome based on electron microscopy and functional studies. The 3D architecture of the primosome reveals an asymmetric, dumbbell-shaped particle. The catalytic centers of primase and Pol α reside in separate lobes of high relative mobility. The flexible tethering of the primosome lobes increases the efficiency of primer transfer between primase and Pol α. The physical organization of the primosome suggests that a concerted mechanism of primer hand-off between primase and Pol α would involve coordinated movements of the primosome lobes. The first three-dimensional map of the eukaryotic primosome at 25 Å resolution provides an essential structural template for understanding initiation of eukaryotic replication.


Assuntos
DNA Polimerase I/química , DNA Polimerase I/ultraestrutura , DNA Primase/química , DNA Primase/ultraestrutura , Sequência de Aminoácidos , DNA Polimerase I/metabolismo , DNA Primase/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Subunidades Proteicas/química , RNA/química , Saccharomyces cerevisiae/enzimologia
20.
Mol Cell ; 34(6): 735-45, 2009 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-19446481

RESUMO

Rad9, Rad1, and Hus1 form a heterotrimeric complex (9-1-1) that is loaded onto DNA at sites of DNA damage. DNA-loaded 9-1-1 activates signaling through the Chk1 arm of the DNA damage checkpoint response via recruitment and stimulation of ATR. Additionally, 9-1-1 may play a direct role in facilitating DNA damage repair via interaction with a number of DNA repair enzymes. We have now determined the crystal structure of the human 9-1-1 complex, revealing a toroidal structure with a similar architecture to the homotrimeric PCNA DNA-binding clamp. The structure explains the formation of a unique heterotrimeric arrangement and reveals significant differences among the three subunits in the sites implicated in binding to the clamp loader and to ligand proteins. Biochemical analysis reveals a single repair enzyme-binding site on 9-1-1 that can be blocked competitively by the PCNA-binding cell-cycle regulator p21(cip1/waf1).


Assuntos
Proteínas de Ciclo Celular/química , Dano ao DNA , Exonucleases/química , Sítios de Ligação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cristalografia por Raios X , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Reparo do DNA , Evolução Molecular , Exonucleases/genética , Exonucleases/metabolismo , Endonucleases Flap/metabolismo , Humanos , Modelos Moleculares , Filogenia , Processamento de Proteína Pós-Traducional , Estrutura Quaternária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...