Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 15(12): e1008228, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31841561

RESUMO

Epstein-Barr virus (EBV) is an oncogenic herpesvirus and WHO class 1 carcinogen that resides in B lymphocytes of nearly all humans. While silent in most, EBV can cause endemic Burkitt lymphoma in children and post-transplant lymphoproliferative disorders/lymphomas in immunocompromised hosts. The pathogenesis of such lymphomas is multifactorial but to a large extent depends on EBV's ability to aggressively drive cellular DNA replication and B cell proliferation despite cell-intrinsic barriers to replication. One such barrier is oncogenic replication stress which hinders the progression of DNA replication forks. To understand how EBV successfully overcomes replication stress, we examined cellular replication forks in EBV-transformed B cells using iPOND (isolation of Proteins on Nascent DNA)-mass spectrometry and identified several cellular proteins that had not previously been linked to DNA replication. Of eight candidate replisome-associated proteins that we validated at forks in EBV-transformed cells and Burkitt lymphoma-derived cells, three zinc finger proteins (ZFPs) were upregulated early in B cells newly-infected with EBV in culture as well as expressed at high levels in EBV-infected B blasts in the blood of immunocompromised transplant recipients. Expressed highly in S- and G2-phase cells, knockdown of each ZFP resulted in stalling of proliferating cells in the S-phase, cleavage of caspase 3, and cell death. These proteins, newly-identified at replication forks of EBV-transformed and Burkitt lymphoma cells therefore contribute to cell survival and cell cycle progression, and represent novel targets for intervention of EBV-lymphomas while simultaneously offering a window into how the replication machinery may be similarly modified in other cancers.


Assuntos
Linfócitos B/virologia , Transformação Celular Viral/fisiologia , Infecções por Vírus Epstein-Barr/metabolismo , Origem de Replicação/fisiologia , Dedos de Zinco/fisiologia , Linfócitos B/patologia , Linfoma de Burkitt/virologia , Proliferação de Células/fisiologia , Herpesvirus Humano 4 , Humanos
2.
BMC Genomics ; 19(1): 623, 2018 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-30134926

RESUMO

BACKGROUND: Genomic regions repressed for DNA replication, resulting in either delayed replication in S phase or underreplication in polyploid cells, are thought to be controlled by inhibition of replication origin activation. Studies in Drosophila polytene cells, however, raised the possibility that impeding replication fork progression also plays a major role. RESULTS: We exploited genomic regions underreplicated (URs) with tissue specificity in Drosophila polytene cells to analyze mechanisms of replication repression. By localizing the Origin Recognition Complex (ORC) in the genome of the larval fat body and comparing this to ORC binding in the salivary gland, we found that sites of ORC binding show extensive tissue specificity. In contrast, there are common domains nearly devoid of ORC in the salivary gland and fat body that also have reduced density of ORC binding sites in diploid cells. Strikingly, domains lacking ORC can still be replicated in some polytene tissues, showing absence of ORC and origins is insufficient to repress replication. Analysis of the width and location of the URs with respect to ORC position indicates that whether or not a genomic region lacking ORC is replicated is controlled by whether replication forks formed outside the region are inhibited. CONCLUSIONS: These studies demonstrate that inhibition of replication fork progression can block replication across genomic regions that constitutively lack ORC. Replication fork progression can be inhibited in both tissue-specific and genome region-specific ways. Consequently, when evaluating sources of genome instability it is important to consider altered control of replication forks in response to differentiation.


Assuntos
Diferenciação Celular/genética , Estruturas Cromossômicas , Replicação do DNA/genética , Organogênese/genética , Complexo de Reconhecimento de Origem/metabolismo , Origem de Replicação/fisiologia , Animais , Sítios de Ligação , Estruturas Cromossômicas/química , Estruturas Cromossômicas/genética , Estruturas Cromossômicas/metabolismo , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Embrião não Mamífero , Larva , Especificidade de Órgãos/genética
3.
Mol Microbiol ; 107(1): 68-80, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29024073

RESUMO

Few studies have described chromosomal dynamics in bacterial cells with more than two complete chromosome copies or described changes with respect to development in polyploid cells. We examined the arrangement of chromosomal loci in the very large, highly polyploid, uncultivated intestinal symbiont Epulopiscium sp. type B using fluorescent in situ hybridization. We found that in new offspring, chromosome replication origins (oriCs) are arranged in a three-dimensional array throughout the cytoplasm. As development progresses, most oriCs become peripherally located. Siblings within a mother cell have similar numbers of oriCs. When chromosome orientation was assessed in situ by labeling two chromosomal regions, no specific pattern was detected. The Epulopiscium genome codes for many of the conserved positional guide proteins used for chromosome segregation in bacteria. Based on this study, we present a model that conserved chromosomal maintenance proteins, combined with entropic demixing, provide the forces necessary for distributing oriCs. Without the positional regulation afforded by radial confinement, chromosomes are more randomly oriented in Epulopiscium than in most small rod-shaped cells. Furthermore, we suggest that the random orientation of individual chromosomes in large polyploid cells would not hamper reproductive success as it would in smaller cells with more limited genomic resources.


Assuntos
Segregação de Cromossomos/fisiologia , Clostridiales/metabolismo , Origem de Replicação/fisiologia , Bactérias/genética , Proteínas de Bactérias/metabolismo , Clostridiales/genética , Replicação do DNA/genética , DNA Bacteriano/metabolismo , Hibridização in Situ Fluorescente , Poliploidia , Origem de Replicação/genética
4.
Biochim Biophys Acta ; 1849(11): 1319-28, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26365772

RESUMO

In metazoans, DNA replication is a highly regulated and ordered process that occurs during the S phase of cell cycle. It begins with the licensing of origins of replication usually found in close proximity of actively transcribing genes owing perhaps to a profound influence of transcription factors on the epigenetic signatures and architecture of chromatin. Here we show that ETS transcription factors are novel regulators of MCM4 origin, whose binding sites are localized between two divergently transcribing MCM4 and PRKDC genes. c-ETS1 and c-ETS2 were recruited to the MCM4 origin respectively during the S and G1 phases of cell cycle. c-ETS2 binding was facilitated by an active chromatin distinguished by acetylated histone H3 orchestrated by histone acetyl transferase GCN5 and followed by HBO1 mediated histone H4 acetylation. Interestingly, c-ETS2 overexpression led to increased BrdU incorporation in the S phase cells while its down-regulation by RNA interference compromised the loading of pre-replicative complex at the origin. Conversely, the recruitment of c-ETS1 at the origin coincided with histone H3 methylation signature characteristic of closed chromatin conformation. As expected, enforced expression of c-ETS1 severely compromised DNA replication whereas its down-regulation enhanced DNA replication as evident from increased BrdU incorporation. Thus, c-ETS transcription factors appear to be key regulators of MCM4 origin where c-ETS2 seems to promote DNA replication whereas c-ETS1 functions as a negative regulator.


Assuntos
Replicação do DNA/fisiologia , Fase G1/fisiologia , Proteína Proto-Oncogênica c-ets-1/metabolismo , Proteína Proto-Oncogênica c-ets-2/metabolismo , Origem de Replicação/fisiologia , Fase S/fisiologia , Proteína Quinase Ativada por DNA/biossíntese , Proteína Quinase Ativada por DNA/genética , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Metilação , Componente 4 do Complexo de Manutenção de Minicromossomo/biossíntese , Componente 4 do Complexo de Manutenção de Minicromossomo/genética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Proteína Proto-Oncogênica c-ets-1/genética , Proteína Proto-Oncogênica c-ets-2/genética
5.
Mol Biol Cell ; 26(23): 4197-208, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26399294

RESUMO

Mitochondrial DNA is prone to damage by various intrinsic as well as environmental stressors. DNA damage can in turn cause problems for replication, resulting in replication stalling and double-strand breaks, which are suspected to be the leading cause of pathological mtDNA rearrangements. In this study, we exposed cells to subtle levels of oxidative stress or UV radiation and followed their effects on mtDNA maintenance. Although the damage did not influence mtDNA copy number, we detected a massive accumulation of RNA:DNA hybrid-containing replication intermediates, followed by an increase in cruciform DNA molecules, as well as in bidirectional replication initiation outside of the main replication origin, OH. Our results suggest that mitochondria maintain two different types of replication as an adaptation to different cellular environments; the RNA:DNA hybrid-involving replication mode maintains mtDNA integrity in tissues with low oxidative stress, and the potentially more error tolerant conventional strand-coupled replication operates when stress is high.


Assuntos
Replicação do DNA/efeitos da radiação , DNA Mitocondrial/metabolismo , Estresse Oxidativo/fisiologia , Células Cultivadas , Variações do Número de Cópias de DNA , Dano ao DNA , Reparo do DNA , Replicação do DNA/fisiologia , DNA Mitocondrial/genética , Relação Dose-Resposta à Radiação , Células HEK293 , Humanos , Hibridização de Ácido Nucleico , Estresse Oxidativo/genética , RNA/genética , RNA/metabolismo , Origem de Replicação/fisiologia , Origem de Replicação/efeitos da radiação , Raios Ultravioleta
6.
Nature ; 519(7544): 431-5, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25739503

RESUMO

Eukaryotic cells initiate DNA replication from multiple origins, which must be tightly regulated to promote precise genome duplication in every cell cycle. To accomplish this, initiation is partitioned into two temporally discrete steps: a double hexameric minichromosome maintenance (MCM) complex is first loaded at replication origins during G1 phase, and then converted to the active CMG (Cdc45-MCM-GINS) helicase during S phase. Here we describe the reconstitution of budding yeast DNA replication initiation with 16 purified replication factors, made from 42 polypeptides. Origin-dependent initiation recapitulates regulation seen in vivo. Cyclin-dependent kinase (CDK) inhibits MCM loading by phosphorylating the origin recognition complex (ORC) and promotes CMG formation by phosphorylating Sld2 and Sld3. Dbf4-dependent kinase (DDK) promotes replication by phosphorylating MCM, and can act either before or after CDK. These experiments define the minimum complement of proteins, protein kinase substrates and co-factors required for regulated eukaryotic DNA replication.


Assuntos
Replicação do DNA , Origem de Replicação/fisiologia , Proteínas de Saccharomyces cerevisiae/isolamento & purificação , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Proteínas de Manutenção de Minicromossomo/metabolismo , Complexos Multienzimáticos/metabolismo , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Origem de Replicação/genética , Proteína de Replicação A/metabolismo , Saccharomyces cerevisiae/enzimologia
7.
Chromosoma ; 124(1): 13-26, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25308420

RESUMO

A crucial step during eukaryotic initiation of DNA replication is the correct loading and activation of the replicative DNA helicase, which ensures that each replication origin fires only once. Unregulated DNA helicase loading and activation, as it occurs in cancer, can cause severe DNA damage and genomic instability. The essential mini-chromosome maintenance proteins 2-7 (MCM2-7) represent the core of the eukaryotic replicative helicase that is loaded at DNA replication origins during G1-phase of the cell cycle. The MCM2-7 helicase activity, however, is only triggered during S-phase once the holo-helicase Cdc45-MCM2-7-GINS (CMG) has been formed. A large number of factors and several kinases interact and contribute to CMG formation and helicase activation, though the exact mechanisms remain unclear. Crucially, upon DNA damage, this reaction is temporarily halted to ensure genome integrity. Here, we review the current understanding of helicase activation; we focus on protein interactions during CMG formation, discuss structural changes during helicase activation, and outline similarities and differences of the prokaryotic and eukaryotic helicase activation process.


Assuntos
Ciclo Celular , Proteínas de Manutenção de Minicromossomo/metabolismo , Origem de Replicação/fisiologia , Ativação Enzimática , Eucariotos/enzimologia , Humanos
8.
Int J Biol Sci ; 10(10): 1193-202, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25516717

RESUMO

SIRT1, the mammalian homolog of yeast Sir2, is a founding member of a family of 7 protein and histone deacetylases that are involved in numerous biological functions. Previous studies revealed that SIRT1 deficiency results in genome instability, which eventually leads to cancer formation, yet the underlying mechanism is unclear. To investigate this, we conducted a proteomics study and found that SIRT1 interacted with many proteins involved in replication fork protection and origin firing. We demonstrated that loss of SIRT1 resulted in increased replication origin firing, asymmetric fork progression, defective intra-S-phase checkpoint, and chromosome damage. Mechanistically, SIRT1 deacetylates and affects the activity of TopBP1, which plays an essential role in DNA replication fork protection and replication origin firing. Our study demonstrated that ectopic over-expression of the deacetylated form of TopBP1 in SIRT1 mutant cells repressed replication origin firing, while the acetylated form of TopBP1 lost this function. Thus, SIRT1 acts upstream of TopBP1 and plays an essential role in maintaining genome stability by modulating DNA replication fork initiation and the intra-S-phase cell cycle checkpoint.


Assuntos
Proteínas de Transporte/metabolismo , Instabilidade Genômica/genética , Origem de Replicação/fisiologia , Pontos de Checagem da Fase S do Ciclo Celular/fisiologia , Sirtuína 1/metabolismo , Acetilação , Animais , Western Blotting , Bromodesoxiuridina , Análise Citogenética , Vetores Genéticos/genética , Células HEK293 , Humanos , Imunoprecipitação , Lentivirus , Espectrometria de Massas , Camundongos , Camundongos Knockout , RNA Interferente Pequeno/genética , Sirtuína 1/genética
9.
Genes Dev ; 28(15): 1653-66, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25085418

RESUMO

The regulated loading of the replicative helicase minichromosome maintenance proteins 2-7 (MCM2-7) onto replication origins is a prerequisite for replication fork establishment and genomic stability. Origin recognition complex (ORC), Cdc6, and Cdt1 assemble two MCM2-7 hexamers into one double hexamer around dsDNA. Although the MCM2-7 hexamer can adopt a ring shape with a gap between Mcm2 and Mcm5, it is unknown which Mcm interface functions as the DNA entry gate during regulated helicase loading. Here, we establish that the Saccharomyces cerevisiae MCM2-7 hexamer assumes a closed ring structure, suggesting that helicase loading requires active ring opening. Using a chemical biology approach, we show that ORC-Cdc6-Cdt1-dependent helicase loading occurs through a unique DNA entry gate comprised of the Mcm2 and Mcm5 subunits. Controlled inhibition of DNA insertion triggers ATPase-driven complex disassembly in vitro, while in vivo analysis establishes that Mcm2/Mcm5 gate opening is essential for both helicase loading onto chromatin and cell cycle progression. Importantly, we demonstrate that the MCM2-7 helicase becomes loaded onto DNA as a single hexamer during ORC/Cdc6/Cdt1/MCM2-7 complex formation prior to MCM2-7 double hexamer formation. Our study establishes the existence of a unique DNA entry gate for regulated helicase loading, revealing key mechanisms in helicase loading, which has important implications for helicase activation.


Assuntos
DNA Fúngico/metabolismo , Proteínas de Manutenção de Minicromossomo/metabolismo , Subunidades Proteicas/metabolismo , Saccharomyces cerevisiae/enzimologia , Trifosfato de Adenosina/metabolismo , Ciclo Celular , Cromossomos Fúngicos/metabolismo , Ativação Enzimática , Hidrólise , Proteínas de Manutenção de Minicromossomo/química , Proteínas de Manutenção de Minicromossomo/genética , Proteínas de Manutenção de Minicromossomo/ultraestrutura , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexos Multiproteicos/ultraestrutura , Subunidades Proteicas/química , Subunidades Proteicas/genética , Origem de Replicação/fisiologia , Saccharomyces cerevisiae/genética
10.
Curr Biol ; 24(3): R111-3, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24502782

RESUMO

Two new studies reveal the main actors involved in the resolution and segregation of newly replicated origins in bacteria. These results have important implications for our understanding of the mechanisms involved in precisely coordinating chromosome organization, segregation and replication.


Assuntos
Adenosina Trifosfatases/metabolismo , Bacillus subtilis/fisiologia , Proteínas de Bactérias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Segregação de Cromossomos/fisiologia , Cromossomos Bacterianos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Mitose/fisiologia , Complexos Multiproteicos/metabolismo , Origem de Replicação/fisiologia
11.
Curr Biol ; 24(3): 287-92, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24440393

RESUMO

SMC condensin complexes play a central role in organizing and compacting chromosomes in all domains of life [1, 2]. In the bacterium Bacillus subtilis, cells lacking SMC are viable only during slow growth and display decondensed chromosomes, suggesting that SMC complexes function throughout the genome [3, 4]. Here, we show that rapid inactivation of SMC or its partner protein ScpB during fast growth leads to a failure to resolve newly replicated origins and a complete block to chromosome segregation. Importantly, the loss of origin segregation is not due to an inability to unlink precatenated sister chromosomes by Topoisomerase IV. In support of the idea that ParB-mediated recruitment of SMC complexes to the origin is important for their segregation, cells with reduced levels of SMC that lack ParB are severely impaired in origin resolution. Finally, we demonstrate that origin segregation is a task shared by the condensin complex and the parABS partitioning system. We propose that origin-localized SMC constrains adjacent DNA segments along their lengths, drawing replicated origins in on themselves and away from each other. This SMC-mediated lengthwise condensation, bolstered by the parABS system, drives origin segregation.


Assuntos
Bacillus subtilis/fisiologia , Proteínas de Bactérias/metabolismo , Proteínas de Ciclo Celular/metabolismo , Segregação de Cromossomos/fisiologia , Mitose/fisiologia , Adenosina Trifosfatases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Microscopia de Fluorescência , Complexos Multiproteicos/metabolismo , Origem de Replicação/fisiologia
12.
Curr Biol ; 24(3): 293-8, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24440399

RESUMO

Condensin-an SMC-kleisin complex-is essential for efficient segregation of sister chromatids in eukaryotes [1-4]. In Escherichia coli and Bacillus subtilis, deletion of condensin subunits results in severe growth phenotypes and the accumulation of cells lacking nucleoids [5, 6]. In many other bacteria and under slow growth conditions, however, the reported phenotypes are much milder or virtually absent [7-10]. This raises the question of what role prokaryotic condensin might play during chromosome segregation under various growth conditions. In B. subtilis and Streptococcus pneumoniae, condensin complexes are enriched on the circular chromosome near the single origin of replication by ParB proteins bound to parS sequences [11, 12]. Using conditional alleles of condensin in B. subtilis, we demonstrate that depletion of its activity results in an immediate and severe defect in the partitioning of replication origins. Multiple copies of the chromosome remain unsegregated at or near the origin of replication. Surprisingly, the growth and chromosome segregation defects in rich medium are suppressed by a reduction of replication fork velocity but not by partial inhibition of translation or transcription. Prokaryotic condensin likely prevents the formation of sister DNA interconnections at the replication fork or promotes their resolution behind the fork.


Assuntos
Adenosina Trifosfatases/metabolismo , Bacillus subtilis/fisiologia , Proteínas de Bactérias/metabolismo , Segregação de Cromossomos/fisiologia , Cromossomos Bacterianos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexos Multiproteicos/metabolismo , Origem de Replicação/fisiologia
13.
Nature ; 492(7428): 205-9, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23201686

RESUMO

Replicative DNA helicases generally unwind DNA as a single hexamer that encircles and translocates along one strand of the duplex while excluding the complementary strand (known as steric exclusion). By contrast, large T antigen, the replicative DNA helicase of the simian virus 40 (SV40), is reported to function as a pair of stacked hexamers that pumps double-stranded DNA through its central channel while laterally extruding single-stranded DNA. Here we use single-molecule and ensemble assays to show that large T antigen assembled on the SV40 origin unwinds DNA efficiently as a single hexamer that translocates on single-stranded DNA in the 3'-to-5' direction. Unexpectedly, large T antigen unwinds DNA past a DNA-protein crosslink on the translocation strand, suggesting that the large T antigen ring can open to bypass bulky adducts. Together, our data underscore the profound conservation among replicative helicase mechanisms, and reveal a new level of plasticity in the interactions of replicative helicases with DNA damage.


Assuntos
DNA Helicases/metabolismo , Vírus 40 dos Símios/enzimologia , Antígenos Virais de Tumores/metabolismo , Replicação do DNA , DNA de Cadeia Simples/metabolismo , DNA Viral/metabolismo , Origem de Replicação/fisiologia , Proteínas Virais/metabolismo
14.
J Virol ; 86(23): 12806-15, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22973044

RESUMO

In human papillomavirus DNA replication, the viral protein E2 forms homodimers and binds to 12-bp palindromic DNA sequences surrounding the origin of DNA replication. Via a protein-protein interaction, it then recruits the viral helicase E1 to an A/T-rich origin of replication, whereupon a dihexamer forms, resulting in DNA replication initiation. In order to carry out DNA replication, the viral proteins must interact with host factors that are currently not all known. An attractive cellular candidate for regulating viral replication is TopBP1, a known interactor of the E2 protein. In mammalian DNA replication, TopBP1 loads DNA polymerases onto the replicative helicase after the G(1)-to-S transition, and this process is tightly cell cycle controlled. The direct interaction between E2 and TopBP1 would allow E2 to bypass this cell cycle control, resulting in DNA replication more than once per cell cycle, which is a requirement for the viral life cycle. We report here the generation of an HPV16 E2 mutant compromised in TopBP1 interaction in vivo and demonstrate that this mutant retains transcriptional activation and repression functions but has suboptimal DNA replication potential. Introduction of this mutant into a viral life cycle model results in the failure to establish viral episomes. The results present a potential new antiviral target, the E2-TopBP1 interaction, and increase our understanding of the viral life cycle, suggesting that the E2-TopBP1 interaction is essential.


Assuntos
Proteínas de Transporte/metabolismo , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Papillomavirus Humano 16/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Southern Blotting , Western Blotting , Primers do DNA/genética , Densitometria , Dimerização , Células HEK293 , Papillomavirus Humano 16/metabolismo , Humanos , Imunoprecipitação , Mutagênese Sítio-Dirigida , Plasmídeos/genética , Origem de Replicação/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Artigo em Inglês | MEDLINE | ID: mdl-22904560

RESUMO

Only ∼10% of replication origins that are licensed by loading minichromosome maintenance 2-7 (MCM2-7) complexes are normally used, with the majority remaining dormant. If replication fork progression is inhibited, nearby dormant origins initiate to ensure that all of the chromosomal DNA is replicated. At the same time, DNA damage-response kinases are activated, which preferentially suppress the assembly of new replication factories. This diverts initiation events away from completely new areas of the genome toward regions experiencing replicative stress. Mice hypomorphic for MCM2-7, which activate fewer dormant origins in response to replication inhibition, are cancer-prone and are genetically unstable. The licensing checkpoint delays entry into S phase if an insufficient number of origins have been licensed. In contrast, humans with Meier-Gorlin syndrome have mutations in pre-RC proteins and show defects in cell proliferation that may be a consequence of chronic activation of the licensing checkpoint.


Assuntos
Replicação do DNA/fisiologia , Modelos Genéticos , Origem de Replicação/fisiologia , Animais , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Microtia Congênita , Dano ao DNA , Orelha/anormalidades , Transtornos do Crescimento/genética , Humanos , Camundongos , Micrognatismo/genética , Patela/anormalidades , Origem de Replicação/genética , Estresse Fisiológico
16.
J Biol Chem ; 286(45): 39200-10, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21937426

RESUMO

From late mitosis to the G(1) phase of the cell cycle, ORC, CDC6, and Cdt1 form the machinery necessary to load MCM2-7 complexes onto DNA. Here, we show that SNF2H, a member of the ATP-dependent chromatin-remodeling complex, is recruited onto DNA replication origins in human cells in a Cdt1-dependent manner and positively regulates MCM loading. SNF2H physically interacted with Cdt1. ChIP assays indicated that SNF2H associates with replication origins specifically during the G(1) phase. Binding of SNF2H at origins was decreased by Cdt1 silencing and, conversely, enhanced by Cdt1 overexpression. Furthermore, SNF2H silencing prevented MCM loading at origins and moderately inhibited S phase progression. Although neither SNF2H overexpression nor SNF2H silencing appeared to impact rereplication induced by Cdt1 overexpression, Cdt1-induced checkpoint activation was inhibited by SNF2H silencing. Collectively, these data suggest that SNF2H may promote MCM loading at DNA replication origins via interaction with Cdt1 in human cells. Because efficient loading of excess MCM complexes is thought to be required for cells to tolerate replication stress, Cdt1- and SNF2H-mediated promotion of MCM loading may be biologically relevant for the regulation of DNA replication.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Replicação do DNA/fisiologia , Fase G1/fisiologia , Complexos Multiproteicos/metabolismo , Origem de Replicação/fisiologia , Fase S/fisiologia , Adenosina Trifosfatases/genética , Proteínas de Ciclo Celular/genética , Montagem e Desmontagem da Cromatina/fisiologia , Proteínas Cromossômicas não Histona/genética , Inativação Gênica , Células HEK293 , Células HeLa , Humanos , Complexos Multiproteicos/genética , Ligação Proteica
17.
J Biol Chem ; 286(33): 29336-29346, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21708944

RESUMO

Chromosomal replication is initiated from the replication origin oriC in Escherichia coli by the active ATP-bound form of DnaA protein. The regulatory inactivation of DnaA (RIDA) system, a complex of the ADP-bound Hda and the DNA-loaded replicase clamp, represses extra initiations by facilitating DnaA-bound ATP hydrolysis, yielding the inactive ADP-bound form of DnaA. However, the mechanisms involved in promoting the DnaA-Hda interaction have not been determined except for the involvement of an interaction between the AAA+ domains of the two. This study revealed that DnaA Leu-422 and Pro-423 residues within DnaA domain IV, including a typical DNA-binding HTH motif, are specifically required for RIDA-dependent ATP hydrolysis in vitro and that these residues support efficient interaction with the DNA-loaded clamp·Hda complex and with Hda in vitro. Consistently, substitutions of these residues caused accumulation of ATP-bound DnaA in vivo and oriC-dependent inhibition of cell growth. Leu-422 plays a more important role in these activities than Pro-423. By contrast, neither of these residues is crucial for DNA replication from oriC, although they are highly conserved in DnaA orthologues. Structural analysis of a DnaA·Hda complex model suggested that these residues make contact with residues in the vicinity of the Hda AAA+ sensor I that participates in formation of a nucleotide-interacting surface. Together, the results show that functional DnaA-Hda interactions require a second interaction site within DnaA domain IV in addition to the AAA+ domain and suggest that these interactions are crucial for the formation of RIDA complexes that are active for DnaA-ATP hydrolysis.


Assuntos
Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/metabolismo , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/metabolismo , Escherichia coli K12/enzimologia , Proteínas de Escherichia coli/metabolismo , Complexos Multienzimáticos/metabolismo , Adenosina Trifosfatases/genética , Trifosfato de Adenosina/genética , Motivos de Aminoácidos , Proteínas de Bactérias/genética , DNA Bacteriano/genética , Proteínas de Ligação a DNA/genética , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Hidrólise , Complexos Multienzimáticos/genética , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Origem de Replicação/fisiologia
18.
J Cell Biol ; 191(7): 1285-97, 2010 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-21173116

RESUMO

Replication origins are licensed by loading MCM2-7 hexamers before entry into S phase. However, only ∼10% of licensed origins are normally used in S phase, with the others remaining dormant. When fork progression is inhibited, dormant origins initiate nearby to ensure that all of the DNA is eventually replicated. In apparent contrast, replicative stress activates ataxia telangiectasia and rad-3-related (ATR) and Chk1 checkpoint kinases that inhibit origin firing. In this study, we show that at low levels of replication stress, ATR/Chk1 predominantly suppresses origin initiation by inhibiting the activation of new replication factories, thereby reducing the number of active factories. At the same time, inhibition of replication fork progression allows dormant origins to initiate within existing replication factories. The inhibition of new factory activation by ATR/Chk1 therefore redirects replication toward active factories where forks are inhibited and away from regions that have yet to start replication. This minimizes the deleterious consequences of fork stalling and prevents similar problems from arising in unreplicated regions of the genome.


Assuntos
Replicação do DNA/fisiologia , Proteínas Quinases/metabolismo , Origem de Replicação/fisiologia , Afidicolina/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia , Bromodesoxiuridina/farmacologia , Cafeína/farmacologia , Carbocianinas/metabolismo , Proteínas de Ciclo Celular/agonistas , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , DNA/metabolismo , Dano ao DNA/fisiologia , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , Nucleotídeos de Desoxiuracil/metabolismo , Fibroblastos/metabolismo , Raios gama , Humanos , Hidroxiureia/farmacologia , Modelos Biológicos , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Purinas/farmacologia , RNA Interferente Pequeno/genética , Origem de Replicação/efeitos dos fármacos , Origem de Replicação/efeitos da radiação , Roscovitina
19.
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
20.
J Proteome Res ; 9(10): 5445-60, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20707412

RESUMO

An origin activation checkpoint has recently been discovered in the G1 phase of the mitotic cell cycle, which can be triggered by loss of DNA replication initiation factors such as the Cdc7 kinase. Insufficient levels of Cdc7 activate cell cycle arrest in normal cells, whereas cancer cells appear to lack this checkpoint response, do not arrest, and proceed with an abortive S phase, leading to cell death. The differential response between normal and tumor cells at this checkpoint has led to widespread interest in the development of pharmacological Cdc7 inhibitors as novel anticancer agents. We have used RNAi against Cdc7 in combination with SILAC-based high resolution MS proteomics to investigate the cellular mechanisms underlying the maintenance of the origin activation checkpoint in normal human diploid fibroblasts. Bioinformatics analysis identified clear changes in wide-ranging biological processes including altered cellular energetic flux, moderate stress response, reduced proliferative capacity, and a spatially distributed response across the mitochondria, lysosomes, and the cell surface. These results provide a quantitative overview of the processes involved in maintenance of the arrested state, show that this phenotype involves active rather than passive cellular adaptation, and highlight a diverse set of proteins responsible for cell cycle arrest and ultimately for promotion of cellular survival. We propose that the Cdc7-depleted proteome maintains cellular arrest by initiating a dynamic quiescence-like response and that the complexities of this phenotype will have important implications for the continued development of promising Cdc7-targeted cancer therapies.


Assuntos
Ciclo Celular/fisiologia , Replicação do DNA/fisiologia , Proteômica/métodos , Origem de Replicação/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Humanos , Immunoblotting , Espectrometria de Massas , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA