Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
PLoS Pathog ; 10(12): e1004536, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25474690

RESUMO

Simian virus 40 (SV40) and cellular DNA replication rely on host ATM and ATR DNA damage signaling kinases to facilitate DNA repair and elicit cell cycle arrest following DNA damage. During SV40 DNA replication, ATM kinase activity prevents concatemerization of the viral genome whereas ATR activity prevents accumulation of aberrant genomes resulting from breakage of a moving replication fork as it converges with a stalled fork. However, the repair pathways that ATM and ATR orchestrate to prevent these aberrant SV40 DNA replication products are unclear. Using two-dimensional gel electrophoresis and Southern blotting, we show that ATR kinase activity, but not DNA-PK(cs) kinase activity, facilitates some aspects of double strand break (DSB) repair when ATM is inhibited during SV40 infection. To clarify which repair factors associate with viral DNA replication centers, we examined the localization of DSB repair proteins in response to SV40 infection. Under normal conditions, viral replication centers exclusively associate with homology-directed repair (HDR) and do not colocalize with non-homologous end joining (NHEJ) factors. Following ATM inhibition, but not ATR inhibition, activated DNA-PK(cs) and KU70/80 accumulate at the viral replication centers while CtIP and BLM, proteins that initiate 5' to 3' end resection during HDR, become undetectable. Similar to what has been observed during cellular DSB repair in S phase, these data suggest that ATM kinase influences DSB repair pathway choice by preventing the recruitment of NHEJ factors to replicating viral DNA. These data may explain how ATM prevents concatemerization of the viral genome and promotes viral propagation. We suggest that inhibitors of DNA damage signaling and DNA repair could be used during infection to disrupt productive viral DNA replication.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Replicação do DNA , DNA Viral/biossíntese , Vírus 40 dos Símios/fisiologia , Replicação Viral/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia/genética , Linhagem Celular , DNA Viral/genética , Humanos
2.
Exp Cell Res ; 334(2): 283-93, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-25933514

RESUMO

The chromosomal DNA replication in eukaryotic cells begins at replication initation sites, which are marked by the assembly of the pre-replication complexes in early G1. At the G1/S transition, recruitment of additional replication initiation proteins enables origin DNA unwinding and loading of DNA polymerases. We found that depletion of the human DNA helicase B (HDHB) inhibits the initiation of DNA replication, suggesting a role of HDHB in the beginning of the DNA synthesis. To gain insight into the function of HDHB during replication initiation, we examined the physical interactions of purified recombinant HDHB with key initiation proteins. HDHB interacts directly with two initiation factors TopBP1 and Cdc45. In addition we found that both, the N-terminus and helicase domain of HDHB bind to the N-terminus of Cdc45. Furthermore depletion of HDHB from human cells diminishes Cdc45 association with chromatin, suggesting that HDHB may facilitate Cdc45 recruitment at G1/S in human cells.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , DNA Helicases/metabolismo , Sítios de Ligação , Proteínas de Ciclo Celular/química , Linhagem Celular , Cromatina/química , DNA Helicases/química , DNA Helicases/deficiência , Humanos
3.
PLoS Pathog ; 9(4): e1003283, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23592994

RESUMO

Mutation of DNA damage checkpoint signaling kinases ataxia telangiectasia-mutated (ATM) or ATM- and Rad3-related (ATR) results in genomic instability disorders. However, it is not well understood how the instability observed in these syndromes relates to DNA replication/repair defects and failed checkpoint control of cell cycling. As a simple model to address this question, we have studied SV40 chromatin replication in infected cells in the presence of inhibitors of ATM and ATR activities. Two-dimensional gel electrophoresis and southern blotting of SV40 chromatin replication products reveal that ATM activity prevents accumulation of unidirectional replication products, implying that ATM promotes repair of replication-associated double strand breaks. ATR activity alleviates breakage of a functional fork as it converges with a stalled fork. The results suggest that during SV40 chromatin replication, endogenous replication stress activates ATM and ATR signaling, orchestrating the assembly of genome maintenance machinery on viral replication intermediates.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Cromatina/metabolismo , Replicação do DNA , Vírus 40 dos Símios/fisiologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/genética , Cafeína/farmacologia , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Chlorocebus aethiops , Dano ao DNA , Reparo do DNA/genética , Replicação do DNA/genética , Humanos , Morfolinas/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Pironas/farmacologia , Vírus 40 dos Símios/genética , Replicação Viral
4.
J Biol Chem ; 287(9): 6469-81, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22194613

RESUMO

Maintenance of genomic stability in proliferating cells depends on a network of proteins that coordinate chromosomal replication with DNA damage responses. Human DNA helicase B (HELB or HDHB) has been implicated in chromosomal replication, but its role in this coordinated network remains undefined. Here we report that cellular exposure to UV irradiation, camptothecin, or hydroxyurea induces accumulation of HDHB on chromatin in a dose- and time-dependent manner, preferentially in S phase cells. Replication stress-induced recruitment of HDHB to chromatin is independent of checkpoint signaling but correlates with the level of replication protein A (RPA) recruited to chromatin. We show using purified proteins that HDHB physically interacts with the N-terminal domain of the RPA 70-kDa subunit (RPA70N). NMR spectroscopy and site-directed mutagenesis reveal that HDHB docks on the same RPA70N surface that recruits S phase checkpoint signaling proteins to chromatin. Consistent with this pattern of recruitment, cells depleted of HDHB display reduced recovery from replication stress.


Assuntos
Dano ao DNA/fisiologia , DNA Helicases/metabolismo , Replicação do DNA/fisiologia , Proteína de Replicação A/metabolismo , Estresse Fisiológico/fisiologia , Sequência de Aminoácidos , Cromossomos/fisiologia , DNA Helicases/química , DNA Helicases/genética , Células HCT116 , Células HeLa , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Osteossarcoma , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas/fisiologia , Proteína de Replicação A/química , Proteína de Replicação A/genética , Pontos de Checagem da Fase S do Ciclo Celular/fisiologia
5.
J Biol Chem ; 287(32): 26854-66, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22700977

RESUMO

DNA polymerase α-primase (Pol-prim) plays an essential role in eukaryotic DNA replication, initiating synthesis of the leading strand and of each Okazaki fragment on the lagging strand. Pol-prim is composed of a primase heterodimer that synthesizes an RNA primer, a DNA polymerase subunit that extends the primer, and a regulatory B-subunit (p68) without apparent enzymatic activity. Pol-prim is thought to interact with eukaryotic replicative helicases, forming a dynamic multiprotein assembly that displays primosome activity. At least three subunits of Pol-prim interact physically with the hexameric replicative helicase SV40 large T antigen, constituting a simple primosome that is active in vitro. However, structural understanding of these interactions and their role in viral chromatin replication in vivo remains incomplete. Here, we report the detailed large T antigen-p68 interface, as revealed in a co-crystal structure and validated by site-directed mutagenesis, and we demonstrate its functional importance in activating the SV40 primosome in cell-free reactions with purified Pol-prim, as well as in monkey cells in vivo.


Assuntos
DNA Polimerase I/metabolismo , DNA Primase/metabolismo , Sequência de Bases , Southern Blotting , DNA Polimerase I/química , DNA Primase/química , Primers do DNA , Replicação do DNA , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Vírus 40 dos Símios/genética
6.
Am J Hum Genet ; 85(5): 606-16, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19853235

RESUMO

We have discovered a distinct DNA-methylation boundary at a site between 650 and 800 nucleotides upstream of the CGG repeat in the first exon of the human FMR1 gene. This boundary, identified by bisulfite sequencing, is present in all human cell lines and cell types, irrespective of age, gender, and developmental stage. The same boundary is found also in different mouse tissues, although sequence homology between human and mouse in this region is only 46.7%. This boundary sequence, in both the unmethylated and the CpG-methylated modes, binds specifically to nuclear proteins from human cells. We interpret this boundary as carrying a specific chromatin structure that delineates a hypermethylated area in the genome from the unmethylated FMR1 promoter and protecting it from the spreading of DNA methylation. In individuals with the fragile X syndrome (FRAXA), the methylation boundary is lost; methylation has penetrated into the FMR1 promoter and inactivated the FMR1 gene. In one FRAXA genome, the upstream terminus of the methylation boundary region exhibits decreased methylation as compared to that of healthy individuals. This finding suggests changes in nucleotide sequence and chromatin structure in the boundary region of this FRAXA individual. In the completely de novo methylated FMR1 promoter, there are isolated unmethylated CpG dinucleotides that are, however, not found when the FMR1 promoter and upstream sequences are methylated in vitro with the bacterial M-SssI DNA methyltransferase. They may arise during de novo methylation only in DNA that is organized in chromatin and be due to the binding of specific proteins.


Assuntos
Metilação de DNA , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Regiões 5' não Traduzidas/genética , Adulto , Animais , Sequência de Bases , Linhagem Celular , Células Cultivadas , Ilhas de CpG , DNA/genética , DNA/isolamento & purificação , Feminino , Fibroblastos/metabolismo , Genoma , Genoma Humano , Células HCT116 , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Análise de Sequência de DNA , Sulfitos/farmacologia
7.
J Biol Chem ; 285(43): 33475-33484, 2010 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-20685648

RESUMO

Replication of simian virus 40 (SV40) DNA, a model for eukaryotic chromosomal replication, can be reconstituted in vitro using the viral helicase (large tumor antigen, or Tag) and purified human proteins. Tag interacts physically with two cellular proteins, replication protein A and DNA polymerase α-primase (pol-prim), constituting the viral primosome. Like the well characterized primosomes of phages T7 and T4, this trio of proteins coordinates parental DNA unwinding with primer synthesis to initiate the leading strand at the viral origin and each Okazaki fragment on the lagging strand template. We recently determined the structure of a previously unrecognized pol-prim domain (p68N) that docks on Tag, identified the p68N surface that contacts Tag, and demonstrated its vital role in primosome function. Here, we identify the p68N-docking site on Tag by using structure-guided mutagenesis of the Tag helicase surface. A charge reverse substitution in Tag disrupted both p68N-binding and primosome activity but did not affect docking with other pol-prim subunits. Unexpectedly, the substitution also disrupted Tag ATPase and helicase activity, suggesting a potential link between p68N docking and ATPase activity. To assess this possibility, we examined the primosome activity of Tag with a single residue substitution in the Walker B motif. Although this substitution abolished ATPase and helicase activity as expected, it did not reduce pol-prim docking on Tag or primosome activity on single-stranded DNA, indicating that Tag ATPase is dispensable for primosome activity in vitro.


Assuntos
Antígenos Transformantes de Poliomavirus/metabolismo , DNA Helicases/metabolismo , DNA Polimerase I/metabolismo , DNA Primase/metabolismo , DNA Viral/metabolismo , Vírus 40 dos Símios/metabolismo , Motivos de Aminoácidos , Antígenos Transformantes de Poliomavirus/genética , DNA/genética , DNA/metabolismo , DNA Helicases/genética , DNA Polimerase I/genética , DNA Primase/genética , Replicação do DNA , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , DNA Viral/genética , Humanos , Mutagênese , Estrutura Terciária de Proteína , Origem de Replicação/fisiologia , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo , Vírus 40 dos Símios/genética
8.
J Biol Chem ; 285(22): 17112-22, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20234039

RESUMO

DNA polymerase alpha-primase (pol-prim) plays a central role in DNA replication in higher eukaryotes, initiating synthesis on both leading and lagging strand single-stranded DNA templates. Pol-prim consists of a primase heterodimer that synthesizes RNA primers, a DNA polymerase that extends them, and a fourth subunit, p68 (also termed B-subunit), that is thought to regulate the complex. Although significant knowledge about single-subunit primases of prokaryotes has accumulated, the functions and regulation of pol-prim remain poorly understood. In the SV40 replication model, the p68 subunit is required for primosome activity and binds directly to the hexameric viral helicase T antigen, suggesting a functional link between T antigen-p68 interaction and primosome activity. To explore this link, we first mapped the interacting regions of the two proteins and discovered a previously unrecognized N-terminal globular domain of p68 (p68N) that physically interacts with the T antigen helicase domain. NMR spectroscopy was used to determine the solution structure of p68N and map its interface with the T antigen helicase domain. Structure-guided mutagenesis of p68 residues in the interface diminished T antigen-p68 interaction, confirming the interaction site. SV40 primosome activity of corresponding pol-prim mutants decreased in proportion to the reduction in p68N-T antigen affinity, confirming that p68-T antigen interaction is vital for primosome function. A model is presented for how this interaction regulates SV40 primosome activity, and the implications of our findings are discussed in regard to the molecular mechanisms of eukaryotic DNA replication initiation.


Assuntos
DNA Polimerase I/química , DNA Primase/química , Vírus 40 dos Símios/enzimologia , Antígenos Virais de Tumores/química , Primers do DNA/genética , Replicação do DNA , Espectroscopia de Ressonância Magnética , Conformação Molecular , Mutagênese , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Técnicas do Sistema de Duplo-Híbrido
9.
Mol Cell Biol ; 27(2): 426-37, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17101793

RESUMO

Fragile X syndrome, the most common form of inherited mental retardation in males, arises when the normally stable 5 to 50 CGG repeats in the 5' untranslated region of the fragile X mental retardation protein 1 (FMR1) gene expand to over 200, leading to DNA methylation and silencing of the FMR1 promoter. Although the events that trigger local CGG expansion remain unknown, the stability of trinucleotide repeat tracts is affected by their position relative to an origin of DNA replication in model systems. Origins of DNA replication in the FMR1 locus have not yet been described. Here, we report an origin of replication adjacent to the FMR1 promoter and CGG repeats that was identified by scanning a 35-kb region. Prereplication proteins Orc3p and Mcm4p bind to chromatin in the FMR1 initiation region in vivo. The position of the FMR1 origin relative to the CGG repeats is consistent with a role in repeat maintenance. The FMR1 origin is active in transformed cell lines, fibroblasts from healthy individuals, fibroblasts from patients with fragile X syndrome, and fetal cells as early as 8 weeks old. The potential role of the FMR1 origin in CGG tract instability is discussed.


Assuntos
Replicação do DNA/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Regiões Promotoras Genéticas , Origem de Replicação , Células Cultivadas , Feminino , Feto/citologia , Fibroblastos/citologia , Humanos , Masculino
10.
Nat Struct Mol Biol ; 12(4): 332-9, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15793585

RESUMO

Simian virus 40 (SV40) provides a model system for the study of eukaryotic DNA replication, in which the viral protein, large T antigen (Tag), marshals human proteins to replicate the viral minichromosome. SV40 replication requires interaction of Tag with the host single-stranded DNA-binding protein, replication protein A (hRPA). The C-terminal domain of the hRPA32 subunit (RPA32C) facilitates initiation of replication, but whether it interacts with Tag is not known. Affinity chromatography and NMR revealed physical interaction between hRPA32C and the Tag origin DNA-binding domain, and a structural model of the complex was determined. Point mutations were then designed to reverse charges in the binding sites, resulting in substantially reduced binding affinity. Corresponding mutations introduced into intact hRPA impaired initiation of replication and primosome activity, implying that this interaction has a critical role in assembly and progression of the SV40 replisome.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Vírus 40 dos Símios/crescimento & desenvolvimento , Replicação Viral/fisiologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Sítios de Ligação , DNA/genética , DNA/metabolismo , Primers do DNA/biossíntese , Primers do DNA/genética , Reparo do DNA , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/imunologia , Humanos , Modelos Moleculares , Mutação/genética , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Estrutura Terciária de Proteína , Proteína de Replicação A , Vírus 40 dos Símios/genética , Replicação Viral/efeitos dos fármacos
11.
J Virol ; 82(11): 5316-28, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18353955

RESUMO

Although the mechanism of simian virus 40 (SV40) DNA replication has been extensively investigated with cell extracts, viral DNA replication in productively infected cells utilizes additional viral and host functions whose interplay remains poorly understood. We show here that in SV40-infected primate cells, the activated ataxia telangiectasia-mutated (ATM) damage-signaling kinase, gamma-H2AX, and Mre11-Rad50-Nbs1 (MRN) assemble with T antigen and other viral DNA replication proteins in large nuclear foci. During infection, steady-state levels of MRN subunits decline, although the corresponding mRNA levels remain unchanged. A proteasome inhibitor stabilizes the MRN complex, suggesting that MRN may undergo proteasome-dependent degradation. Analysis of mutant T antigens with disrupted binding to the ubiquitin ligase CUL7 revealed that MRN subunits are stable in cells infected with mutant virus or transfected with mutant viral DNA, implicating CUL7 association with T antigen in MRN proteolysis. The mutant genomes produce fewer virus progeny than the wild type, suggesting that T antigen-CUL7-directed proteolysis facilitates virus propagation. Use of a specific ATM kinase inhibitor showed that ATM kinase signaling is a prerequisite for proteasome-dependent degradation of MRN subunits as well as for the localization of T antigen and damage-signaling proteins to viral replication foci and optimal viral DNA replication. Taken together, the results indicate that SV40 infection manipulates host DNA damage-signaling to reprogram the cell for viral replication, perhaps through mechanisms related to host recovery from DNA damage.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Vírus 40 dos Símios/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Dano ao DNA/genética , Humanos , Camundongos , Dados de Sequência Molecular , Subunidades Proteicas/metabolismo , Alinhamento de Sequência , Vírus 40 dos Símios/genética , Replicação Viral
12.
Virus Res ; 141(1): 71-80, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19200446

RESUMO

Polyomaviruses such as BK virus and JC virus have been linked to several diseases, but treatments that thwart their propagation are limited in part because of slow growth and cumbersome culturing conditions. In contrast, the replication of one member of this family, Simian Virus 40 (SV40), is robust and has been well-characterized. SV40 replication requires two domains within the viral-encoded large tumor antigen (TAg): The ATPase domain and the N-terminal J domain, which stimulates the ATPase activity of the Hsp70 chaperone. To assess whether inhibitors of polyomavirus replication could be identified, we examined a recently described library of small molecules, some of which inhibit chaperone function. One compound, MAL2-11B, inhibited both TAg's endogenous ATPase activity and the TAg-mediated activation of Hsp70. MAL2-11B also reduced SV40 propagation in plaque assays and compromised DNA replication in cell culture and in vitro. Furthermore, the compound significantly reduced the growth of BK virus in a human kidney cell line. These data indicate that pharmacological inhibition of TAg's chaperone and ATPase activities may provide a route to combat polyomavirus-mediated disease.


Assuntos
Adenosina Trifosfatases/metabolismo , Antígenos Virais de Tumores/metabolismo , Regulação para Baixo , Proteínas de Choque Térmico HSP70/metabolismo , Vírus 40 dos Símios/fisiologia , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Adenosina Trifosfatases/genética , Antígenos Virais de Tumores/genética , Linhagem Celular , Proteínas de Choque Térmico HSP70/genética , Humanos , Vírus 40 dos Símios/efeitos dos fármacos , Vírus 40 dos Símios/genética , Proteínas Virais/genética
14.
Biochem J ; 407(2): 313-20, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17666013

RESUMO

The initiation of SV40 (simian virus 40) DNA replication requires the co-operative interactions between the viral Tag (large T-antigen), RPA (replication protein A) and Pol (DNA polymerase alpha-primase) on the template DNA. Binding interfaces mapped on these enzymes and expressed as peptides competed with the mutual interactions of the native proteins. Prevention of the genuine interactions was accomplished only prior to the primer synthesis step and blocked the assembly of a productive initiation complex. Once the complex was engaged in the synthesis of an RNA primer and its extension, the interfering effects of the peptides ceased, suggesting a stable association of the replication factors during the initiation phase. Specific antibodies were still able to disrupt preformed interactions and inhibited primer synthesis and extension activities, underlining the crucial role of specific protein-protein contacts during the entire initiation process.


Assuntos
Replicação do DNA , Vírus 40 dos Símios/fisiologia , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/metabolismo , Produtos do Gene pol/genética , Produtos do Gene pol/metabolismo , Complexos Multiproteicos/metabolismo , Ligação Proteica , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo , Fatores de Tempo , Proteínas Virais/genética , Proteínas Virais/metabolismo
15.
Nucleic Acids Res ; 34(15): 4126-37, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16935876

RESUMO

Processing of DNA in replication, repair and recombination pathways in cells of all organisms requires the participation of at least one major single-stranded DNA (ssDNA)-binding protein. This protein protects ssDNA from nucleolytic damage, prevents hairpin formation and blocks DNA reannealing until the processing pathway is successfully completed. Many ssDNA-binding proteins interact physically and functionally with a variety of other DNA processing proteins. These interactions are thought to temporally order and guide the parade of proteins that 'trade places' on the ssDNA, a model known as 'hand-off', as the processing pathway progresses. How this hand-off mechanism works remains poorly understood. Recent studies of the conserved eukaryotic ssDNA-binding protein replication protein A (RPA) suggest a novel mechanism by which proteins may trade places on ssDNA by binding to RPA and mediating conformation changes that alter the ssDNA-binding properties of RPA. This article reviews the structure and function of RPA, summarizes recent studies of RPA in DNA replication and other DNA processing pathways, and proposes a general model for the role of RPA in protein-mediated hand-off.


Assuntos
Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteína de Replicação A/fisiologia , Saccharomyces cerevisiae/genética , DNA/metabolismo , DNA de Cadeia Simples/metabolismo , Modelos Biológicos , Conformação Proteica , Proteína de Replicação A/química
16.
FEBS Lett ; 581(21): 3973-8, 2007 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-17673209

RESUMO

Replication protein A (RPA) is a stable heterotrimeric complex consisting of p70, p32 and p14 subunits. The protein plays a crucial role in SV40 minichromosome replication. Peptides of p70 representing interaction sites for the smaller two subunits, DNA as well as the viral initiator protein large T-antigen (Tag) and the cellular DNA polymerase alpha-primase (Pol) all interfered with the replication process indicating the importance of the different p70 activities in this process. Inhibition by the peptide disrupting protein-protein interactions was observed only during the pre-initiation stage prior to primer synthesis, suggesting the formation of a stable initiation complex between RPA, Tag and Pol at the primer end.


Assuntos
Replicação do DNA/fisiologia , DNA Viral/metabolismo , Proteína de Replicação A/metabolismo , Vírus 40 dos Símios/fisiologia , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/metabolismo , Linhagem Celular , DNA Polimerase I/genética , DNA Polimerase I/metabolismo , DNA Primase/genética , DNA Primase/metabolismo , Primers do DNA/genética , Primers do DNA/metabolismo , DNA Viral/genética , Humanos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Ligação Proteica/fisiologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteína de Replicação A/genética , Proteínas Virais/genética
17.
J Mol Biol ; 357(4): 1295-305, 2006 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-16481006

RESUMO

Large T antigen (LTag) from simian virus 40 (SV40) is an ATP-driven DNA helicase that specifically recognizes the core of the viral origin of replication (ori), where it oligomerizes as a double hexamer. During this process, binding of the first hexamer stimulates the assembly of a second one. Using electron microscopy, we show that the N-terminal part of LTag that includes the origin-binding domain does not present a stable quaternary structure in single hexamers. This disordered region, however, is well arranged within the LTag double hexamer after specific ori recognition, where it mediates the interactions between hexamers and constructs a separated structural module at their junction. We conclude that full assembly of LTag hexamers occurs only within the dodecamer, and requires the specific hexamer-hexamer interactions established upon binding to the origin of replication. This mechanism provides the structural basis for the cooperative assembly of LTag double hexamer on the cognate viral ori.


Assuntos
Antígenos Transformantes de Poliomavirus/química , DNA Viral , Estrutura Quaternária de Proteína , Origem de Replicação , Antígenos Transformantes de Poliomavirus/metabolismo , Microscopia Crioeletrônica , Modelos Moleculares , Vírus 40 dos Símios
18.
Mol Cell Biol ; 24(10): 4138-50, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15121836

RESUMO

A small DNA fragment containing the high-frequency initiation region (IR) ori-beta from the hamster dihydrofolate reductase locus functions as an independent replicator in ectopic locations in both hamster and human cells. Conversely, a fragment of the human lamin B2 locus containing the previously mapped IR serves as an independent replicator at ectopic chromosomal sites in hamster cells. At least four defined sequence elements are specifically required for full activity of ectopic ori-beta in hamster cells. These include an AT-rich element, a 4-bp sequence located within the mapped IR, a region of intrinsically bent DNA located between these two elements, and a RIP60 protein binding site adjacent to the bent region. The ori-beta AT-rich element is critical for initiation activity in human, as well as hamster, cells and can be functionally substituted for by an AT-rich region from the human lamin B2 IR that differs in nucleotide sequence and length. Taken together, the results demonstrate that two mammalian replicators can be activated at ectopic sites in chromosomes of another mammal and lead us to speculate that they may share functionally related elements.


Assuntos
Origem de Replicação , Animais , Composição de Bases , Sequência de Bases , Células CHO , Linhagem Celular , Cromossomos/genética , Cromossomos Humanos/genética , Sequência Conservada , Cricetinae , DNA/química , DNA/genética , Replicação do DNA/genética , Células HeLa , Humanos , Lamina Tipo B/genética , Dados de Sequência Molecular , Especificidade da Espécie , Tetra-Hidrofolato Desidrogenase/genética
19.
Mol Cell Biol ; 22(16): 5669-78, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12138179

RESUMO

DNA polymerase alpha-primase (pol-prim) is a heterotetramer with DNA polymerase and primase activities. The polymerase (p180) and primase (p48 and p58) subunits synthesize primers and extend them, but the function of the remaining subunit (p68) is poorly understood. Genetic studies in yeast suggested an essential role for the p68 ortholog in early S phase prior to the hydroxyurea-sensitive step, possibly a regulatory role in initiation of DNA replication, but found no evidence for an essential function of p68 later in S phase. To investigate whether the human p68 subunit has an essential role in DNA replication, we examined the ability of a purified trimeric human pol-prim lacking p68 to initiate simian virus 40 DNA replication in vitro and to synthesize and elongate primers on single-stranded DNA in the presence of T antigen and replication protein A (RPA). Both activities of trimeric pol-prim were defective, but activity was recovered upon addition of separately purified p68. Phosphorylation of p68 by cyclin A-dependent protein kinase also inhibited both activities of pol-prim. The data strongly suggest that the p68 subunit is required for priming activity of pol-prim in the presence of RPA and T antigen, both during initiation at the origin and during lagging strand replication.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , DNA Polimerase I/metabolismo , DNA Primase/metabolismo , Replicação do DNA , Vírus 40 dos Símios/genética , Animais , Antígenos Transformantes de Poliomavirus/metabolismo , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , DNA Polimerase I/genética , DNA Primase/genética , Humanos , Substâncias Macromoleculares , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Quaternária de Proteína , Subunidades Proteicas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vírus 40 dos Símios/imunologia , Vírus 40 dos Símios/fisiologia , Moldes Genéticos , Replicação Viral
20.
Mol Biol Cell ; 15(7): 3320-32, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15146062

RESUMO

Mutational studies of human DNA helicase B (HDHB) have suggested that its activity is critical for the G1/S transition of the cell cycle, but the nature of its role remains unknown. In this study, we show that during G1, ectopically expressed HDHB localizes in nuclear foci induced by DNA damaging agents and that this focal pattern requires active HDHB. During S and G2/M, HDHB localizes primarily in the cytoplasm. A carboxy-terminal domain from HDHB confers cell cycle-dependent localization, but not the focal pattern, to a reporter protein. A cluster of potential cyclin-dependent kinase phosphorylation sites in this domain was modified at the G1/S transition and maintained through G2/M of the cell cycle in vivo, coincident with nuclear export of HDHB. Serine 967 of HDHB was the major site phosphorylated in vivo and in vitro by cyclin-dependent kinases. Mutational analysis demonstrated that phosphorylation of serine 967 is crucial in regulating the subcellular localization of ectopically expressed HDHB. We propose that the helicase of HDHB operates primarily during G1 to process endogenous DNA damage before the G1/S transition, and it is largely sequestered in the cytoplasm during S/G2.


Assuntos
Ciclo Celular , Dano ao DNA , DNA Helicases/análise , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Quinases relacionadas a CDC2 e CDC28/análise , Quinases relacionadas a CDC2 e CDC28/metabolismo , Camptotecina/farmacologia , Quinase 2 Dependente de Ciclina , DNA/efeitos dos fármacos , DNA Helicases/genética , DNA Helicases/metabolismo , Análise Mutacional de DNA , Fase G1/fisiologia , Humanos , Espaço Intracelular/imunologia , Espaço Intracelular/ultraestrutura , Mitomicina/farmacologia , Dados de Sequência Molecular , Fosforilação , Serina/genética , Serina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA