Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Exp Cell Res ; 334(2): 283-93, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25933514

ABSTRACT

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.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/metabolism , DNA Helicases/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , Cell Line , Chromatin/chemistry , DNA Helicases/chemistry , DNA Helicases/deficiency , Humans
2.
PLoS One ; 10(2): e0116093, 2015.
Article in English | MEDLINE | ID: mdl-25706313

ABSTRACT

Simian virus 40 (SV40) serves as an important model organism for studying eukaryotic DNA replication. Its helicase, Large T-antigen (Tag), is a multi-functional protein that interacts with multiple host proteins, including the ubiquitous ssDNA binding protein Replication Protein A (RPA). Tag recruits RPA, actively loads it onto the unwound DNA, and together they promote priming of the template. Although interactions of Tag with RPA have been mapped, no interaction between Tag and the N-terminal protein interaction domain of the RPA 70kDa subunit (RPA70N) has been reported. Here we provide evidence of direct physical interaction of Tag with RPA70N and map the binding sites using a series of pull-down and mutational experiments. In addition, a monoclonal anti-Tag antibody, the epitope of which overlaps with the binding site, blocks the binding of Tag to RPA70N. We use NMR chemical shift perturbation analysis to show that Tag uses the same basic cleft in RPA70N as multiple of DNA damage response proteins. Mutations in the binding sites of both RPA70N and Tag demonstrate that specific charge reversal substitutions in either binding partner strongly diminish the interaction. These results expand the known repertoire of contacts between Tag and RPA, which mediate the many critical roles of Tag in viral replication.


Subject(s)
Antigens, Viral, Tumor/metabolism , DNA Helicases/metabolism , DNA, Viral , Replication Protein A/metabolism , Simian virus 40/immunology , DNA Replication/physiology , Virus Replication/physiology
3.
PLoS One ; 10(1): e0116852, 2015.
Article in English | MEDLINE | ID: mdl-25617833

ABSTRACT

Homologous recombination is involved in the repair of DNA damage and collapsed replication fork, and is critical for the maintenance of genomic stability. Its process involves a network of proteins with different enzymatic activities. Human DNA helicase B (HDHB) is a robust 5'-3' DNA helicase which accumulates on chromatin in cells exposed to DNA damage. HDHB facilitates cellular recovery from replication stress, but its role in DNA damage response remains unclear. Here we report that HDHB silencing results in reduced sister chromatid exchange, impaired homologous recombination repair, and delayed RPA late-stage foci formation induced by ionizing radiation. Ectopically expressed HDHB colocalizes with Rad51, Rad52, RPA, and ssDNA. In vitro, HDHB stimulates Rad51-mediated heteroduplex extension in 5'-3' direction. A helicase-defective mutant HDHB failed to promote this reaction. Our studies implicate HDHB promotes homologous recombination in vivo and stimulates 5'-3' heteroduplex extension during Rad51-mediated strand exchange in vitro.


Subject(s)
DNA Helicases/metabolism , DNA/genetics , Homologous Recombination , Nucleic Acid Heteroduplexes/genetics , Rad51 Recombinase/metabolism , Cell Line, Tumor , Chromatids/genetics , Chromatids/radiation effects , DNA Damage , DNA Repair/radiation effects , DNA, Single-Stranded/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Homologous Recombination/radiation effects , Humans , Protein Transport/radiation effects , Rad52 DNA Repair and Recombination Protein/metabolism , Replication Protein A/metabolism
4.
PLoS Pathog ; 10(12): e1004536, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25474690

ABSTRACT

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.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Replication , DNA, Viral/biosynthesis , Simian virus 40/physiology , Virus Replication/physiology , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line , DNA, Viral/genetics , Humans
5.
J Mol Biol ; 426(3): 558-69, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24239947

ABSTRACT

DNA replication in all organisms requires polymerases to synthesize copies of the genome. DNA polymerases are unable to function on a bare template and require a primer. Primases are crucial RNA polymerases that perform the initial de novo synthesis, generating the first 8-10 nucleotides of the primer. Although structures of archaeal and bacterial primases have provided insights into general priming mechanisms, these proteins are not well conserved with heterodimeric (p48/p58) primases in eukaryotes. Here, we present X-ray crystal structures of the catalytic engine of a eukaryotic primase, which is contained in the p48 subunit. The structures of p48 reveal that eukaryotic primases maintain the conserved catalytic prim fold domain, but with a unique subdomain not found in the archaeal and bacterial primases. Calorimetry experiments reveal that Mn(2+) but not Mg(2+) significantly enhances the binding of nucleotide to primase, which correlates with higher catalytic efficiency in vitro. The structure of p48 with bound UTP and Mn(2+) provides insights into the mechanism of nucleotide synthesis by primase. Substitution of conserved residues involved in either metal or nucleotide binding alter nucleotide binding affinities, and yeast strains containing the corresponding Pri1p substitutions are not viable. Our results reveal that two residues (S160 and H166) in direct contact with the nucleotide were previously unrecognized as critical to the human primase active site. Comparing p48 structures to those of similar polymerases in different states of action suggests changes that would be required to attain a catalytically competent conformation capable of initiating dinucleotide synthesis.


Subject(s)
DNA Primase/chemistry , DNA Primers/chemical synthesis , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , DNA Primase/metabolism , Humans , Manganese/metabolism , Protein Conformation , Protein Subunits , Saccharomyces cerevisiae/metabolism
6.
PLoS Pathog ; 9(4): e1003283, 2013.
Article in English | MEDLINE | ID: mdl-23592994

ABSTRACT

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.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Chromatin/metabolism , DNA Replication , Simian virus 40/physiology , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Caffeine/pharmacology , Cell Cycle Checkpoints , Cell Line , Chlorocebus aethiops , DNA Damage , DNA Repair/genetics , DNA Replication/genetics , Humans , Morpholines/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Pyrones/pharmacology , Simian virus 40/genetics , Virus Replication
8.
J Biol Chem ; 287(32): 26854-66, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22700977

ABSTRACT

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.


Subject(s)
DNA Polymerase I/metabolism , DNA Primase/metabolism , Base Sequence , Blotting, Southern , DNA Polymerase I/chemistry , DNA Primase/chemistry , DNA Primers , DNA Replication , Humans , Models, Molecular , Molecular Sequence Data , Simian virus 40/genetics
9.
J Biol Chem ; 287(9): 6469-81, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22194613

ABSTRACT

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.


Subject(s)
DNA Damage/physiology , DNA Helicases/metabolism , DNA Replication/physiology , Replication Protein A/metabolism , Stress, Physiological/physiology , Amino Acid Sequence , Chromosomes/physiology , DNA Helicases/chemistry , DNA Helicases/genetics , HCT116 Cells , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Osteosarcoma , Protein Folding , Protein Interaction Domains and Motifs/physiology , Replication Protein A/chemistry , Replication Protein A/genetics , S Phase Cell Cycle Checkpoints/physiology
11.
J Biol Chem ; 285(43): 33475-33484, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20685648

ABSTRACT

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.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , DNA Helicases/metabolism , DNA Polymerase I/metabolism , DNA Primase/metabolism , DNA, Viral/metabolism , Simian virus 40/metabolism , Amino Acid Motifs , Antigens, Polyomavirus Transforming/genetics , DNA/genetics , DNA/metabolism , DNA Helicases/genetics , DNA Polymerase I/genetics , DNA Primase/genetics , DNA Replication , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA, Viral/genetics , Humans , Mutagenesis , Protein Structure, Tertiary , Replication Origin/physiology , Replication Protein A/genetics , Replication Protein A/metabolism , Simian virus 40/genetics
14.
J Biol Chem ; 285(22): 17112-22, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20234039

ABSTRACT

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.


Subject(s)
DNA Polymerase I/chemistry , DNA Primase/chemistry , Simian virus 40/enzymology , Antigens, Viral, Tumor/chemistry , DNA Primers/genetics , DNA Replication , Magnetic Resonance Spectroscopy , Molecular Conformation , Mutagenesis , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Two-Hybrid System Techniques
15.
J Mol Biol ; 397(5): 1276-86, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20219473

ABSTRACT

The Simian virus 40 (SV40) large tumor antigen (LTag) functions as the replicative helicase and initiator for viral DNA replication. For SV40 replication, the first essential step is the assembly of an LTag double hexamer at the origin DNA that will subsequently melt the origin DNA to initiate fork unwinding. In this study, we used three-dimensional cryo-electron microscopy to visualize early events in the activation of DNA replication in the SV40 model system. We obtained structures of wild-type double-hexamer complexes of LTag bound to SV40 origin DNA, to which atomic structures have been fitted. Wild-type LTag was observed in two distinct conformations: In one conformation, the central module containing the J-domains and the origin binding domains of both hexamers is a compact closed ring. In the other conformation, the central module is an open ring with a gap formed by rearrangement of the N-terminal regions of the two hexamers, potentially allowing for the passage of single-stranded DNA generated from the melted origin DNA. Double-hexamer complexes containing mutant LTag that lacks the N-terminal J-domain show the central module predominantly in the closed-ring state. Analyses of the LTag C-terminal regions reveal that the LTag hexamers bound to the A/T-rich tract origin of replication and early palindrome origin of replication elements are structurally distinct. Lastly, visualization of DNA density protruding from the LTag C-terminal domains suggests that oligomerization of the LTag complex takes place on double-stranded DNA.


Subject(s)
Antigens, Viral, Tumor/chemistry , Simian virus 40/genetics , Virus Replication/genetics , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/metabolism , Binding Sites , Cryoelectron Microscopy , DNA Replication/genetics , DNA, Viral/metabolism , DNA, Viral/physiology , Protein Conformation , Protein Multimerization , Simian virus 40/chemistry
16.
Am J Hum Genet ; 85(5): 606-16, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19853235

ABSTRACT

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.


Subject(s)
DNA Methylation , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , 5' Untranslated Regions/genetics , Adult , Animals , Base Sequence , Cell Line , Cells, Cultured , CpG Islands , DNA/genetics , DNA/isolation & purification , Female , Fibroblasts/metabolism , Genome , Genome, Human , HCT116 Cells , Humans , Male , Mice , Molecular Sequence Data , Protein Binding , Sequence Analysis, DNA , Sulfites/pharmacology
17.
J Biol Chem ; 284(36): 24662-72, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19608746

ABSTRACT

Mcm10 is an essential eukaryotic protein required for the initiation and elongation phases of chromosomal replication. Specifically, Mcm10 is required for the association of several replication proteins, including DNA polymerase alpha (pol alpha), with chromatin. We showed previously that the internal (ID) and C-terminal (CTD) domains of Mcm10 physically interact with both single-stranded (ss) DNA and the catalytic p180 subunit of pol alpha. However, the mechanism by which Mcm10 interacts with pol alpha on and off DNA is unclear. As a first step toward understanding the structural details for these critical intermolecular interactions, x-ray crystallography and NMR spectroscopy were used to map the binary interfaces between Mcm10-ID, ssDNA, and p180. The crystal structure of an Mcm10-ID*ssDNA complex confirmed and extended our previous evidence that ssDNA binds within the oligonucleotide/oligosaccharide binding-fold cleft of Mcm10-ID. We show using NMR chemical shift perturbation and fluorescence spectroscopy that p180 also binds to the OB-fold and that ssDNA and p180 compete for binding to this motif. In addition, we map a minimal Mcm10 binding site on p180 to a small region within the p180 N-terminal domain (residues 286-310). These findings, together with data for DNA and p180 binding to an Mcm10 construct that contains both the ID and CTD, provide the first mechanistic insight into how Mcm10 might use a handoff mechanism to load and stabilize pol alpha within the replication fork.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Polymerase I/chemistry , DNA, Single-Stranded/chemistry , Amino Acid Motifs/physiology , Binding Sites/physiology , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Crystallography, X-Ray , DNA Polymerase I/genetics , DNA Polymerase I/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Humans , Minichromosome Maintenance Proteins , Nuclear Magnetic Resonance, Biomolecular , Protein Binding/physiology
18.
Virus Res ; 141(1): 71-80, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19200446

ABSTRACT

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.


Subject(s)
Adenosine Triphosphatases/metabolism , Antigens, Viral, Tumor/metabolism , Down-Regulation , HSP70 Heat-Shock Proteins/metabolism , Simian virus 40/physiology , Small Molecule Libraries/pharmacology , Viral Proteins/metabolism , Virus Replication/drug effects , Adenosine Triphosphatases/genetics , Antigens, Viral, Tumor/genetics , Cell Line , HSP70 Heat-Shock Proteins/genetics , Humans , Simian virus 40/drug effects , Simian virus 40/genetics , Viral Proteins/genetics
19.
Virology ; 384(2): 352-9, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19101707

ABSTRACT

Duplication of the simian virus 40 (SV40) genome is the best understood eukaryotic DNA replication process to date. Like most prokaryotic genomes, the SV40 genome is a circular duplex DNA organized in a single replicon. This small viral genome, its association with host histones in nucleosomes, and its dependence on the host cell milieu for replication factors and precursors led to its adoption as a simple and powerful model. The steps in replication, the viral initiator, the host proteins, and their mechanisms of action were initially defined using a cell-free SV40 replication reaction. Although our understanding of the vastly more complex host replication fork is advancing, no eukaryotic replisome has yet been reconstituted and the SV40 paradigm remains a point of reference. This article reviews some of the milestones in the development of this paradigm and speculates on its potential utility to address unsolved questions in eukaryotic genome maintenance.


Subject(s)
DNA Replication , Simian virus 40/genetics , Virus Replication , Antigens, Polyomavirus Transforming/metabolism , DNA, Viral/genetics , Genome, Viral , Simian virus 40/metabolism , Simian virus 40/physiology
20.
J Virol ; 82(11): 5316-28, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18353955

ABSTRACT

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.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Kinases/metabolism , Signal Transduction , Simian virus 40/metabolism , Amino Acid Sequence , Animals , Cell Line , DNA Damage/genetics , Humans , Mice , Molecular Sequence Data , Protein Subunits/metabolism , Sequence Alignment , Simian virus 40/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL