Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nicholas P. George is active.

Publication


Featured researches published by Nicholas P. George.


The EMBO Journal | 2011

Structure of the SSB–DNA polymerase III interface and its role in DNA replication

Aimee H. Marceau; Soon Bahng; Shawn C. Massoni; Nicholas P. George; Steven J. Sandler; Kenneth J. Marians; James L. Keck

Interactions between single‐stranded DNA‐binding proteins (SSBs) and the DNA replication machinery are found in all organisms, but the roles of these contacts remain poorly defined. In Escherichia coli, SSBs association with the χ subunit of the DNA polymerase III holoenzyme has been proposed to confer stability to the replisome and to aid delivery of primers to the lagging‐strand DNA polymerase. Here, the SSB‐binding site on χ is identified crystallographically and biochemical and cellular studies delineate the consequences of destabilizing the χ/SSB interface. An essential role for the χ/SSB interaction in lagging‐strand primer utilization is not supported. However, sequence changes in χ that block complex formation with SSB lead to salt‐dependent uncoupling of leading‐ and lagging‐strand DNA synthesis and to a surprising obstruction of the leading‐strand DNA polymerase in vitro, pointing to roles for the χ/SSB complex in replisome establishment and maintenance. Destabilization of the χ/SSB complex in vivo produces cells with temperature‐dependent cell cycle defects that appear to arise from replisome instability.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Structural mechanisms of PriA-mediated DNA replication restart.

Basudeb Bhattacharyya; Nicholas P. George; Tiffany Thurmes; Ruobo Zhou; Niketa Jani; Sarah R. Wessel; Steven J. Sandler; Taekjip Ha; James L. Keck

Significance This study describes the crystal structures of the full-length PriA DNA helicase, a multifunctional enzyme that mediates the essential process of restarting prematurely terminated DNA replication reactions in bacteria. Our findings reveal how PriA is able to recognize replication restart substrates through structure-specific DNA binding and interactions with the ssDNA-binding protein and how it exposes ssDNA that could be used to reload the replisome and reinitiate replication. These observations shed light on the physical mechanisms that allow cells to survive the common and potentially lethal problems posed by incomplete genome replication. Collisions between cellular DNA replication machinery (replisomes) and damaged DNA or immovable protein complexes can dissociate replisomes before the completion of replication. This potentially lethal problem is resolved by cellular “replication restart” reactions that recognize the structures of prematurely abandoned replication forks and mediate replisomal reloading. In bacteria, this essential activity is orchestrated by the PriA DNA helicase, which identifies replication forks via structure-specific DNA binding and interactions with fork-associated ssDNA-binding proteins (SSBs). However, the mechanisms by which PriA binds replication fork DNA and coordinates subsequent replication restart reactions have remained unclear due to the dearth of high-resolution structural information available for the protein. Here, we describe the crystal structures of full-length PriA and PriA bound to SSB. The structures reveal a modular arrangement for PriA in which several DNA-binding domains surround its helicase core in a manner that appears to be poised for binding to branched replication fork DNA structures while simultaneously allowing complex formation with SSB. PriA interaction with SSB is shown to modulate SSB/DNA complexes in a manner that exposes a potential replication initiation site. From these observations, a model emerges to explain how PriA links recognition of diverse replication forks to replication restart.


Nucleic Acids Research | 2012

Genetic recombination in Bacillus subtilis: a division of labor between two single-strand DNA-binding proteins

Tribhuwan Yadav; Begoña Carrasco; Angela R. Myers; Nicholas P. George; James L. Keck; Juan Carlos Alonso

We have investigated the structural, biochemical and cellular roles of the two single-stranded (ss) DNA-binding proteins from Bacillus subtilis, SsbA and SsbB. During transformation, SsbB localizes at the DNA entry pole where it binds and protects internalized ssDNA. The 2.8-Å resolution structure of SsbB bound to ssDNA reveals a similar overall protein architecture and ssDNA-binding surface to that of Escherichia coli SSB. SsbA, which binds ssDNA with higher affinity than SsbB, co-assembles onto SsbB-coated ssDNA and the two proteins inhibit ssDNA binding by the recombinase RecA. During chromosomal transformation, the RecA mediators RecO and DprA provide RecA access to ssDNA. Interestingly, RecO interaction with ssDNA-bound SsbA helps to dislodge both SsbA and SsbB from the DNA more efficiently than if the DNA is coated only with SsbA. Once RecA is nucleated onto the ssDNA, RecA filament elongation displaces SsbA and SsbB and enables RecA-mediated DNA strand exchange. During plasmid transformation, RecO localizes to the entry pole and catalyzes annealing of SsbA- or SsbA/SsbB-coated complementary ssDNAs to form duplex DNA with ssDNA tails. Our results provide a mechanistic framework for rationalizing the coordinated events modulated by SsbA, SsbB and RecO that are crucial for RecA-dependent chromosomal transformation and RecA-independent plasmid transformation.


Journal of Biological Chemistry | 2012

Structure and Cellular Dynamics of Deinococcus radiodurans Single-stranded DNA (ssDNA)-binding Protein (SSB)-DNA Complexes

Nicholas P. George; Khanh V. Ngo; Sindhu Chitteni-Pattu; Cédric Norais; John R. Battista; Michael M. Cox; James L. Keck

Background: The dimeric D. radiodurans single-stranded DNA-binding protein (DrSSB) has poorly defined DNA-binding and cellular mechanisms. Results: DrSSB binds ssDNA analogously to tetrameric bacterial SSBs and is regulated in response to ionizing radiation. Conclusion: The DrSSB ssDNA mechanism is conserved with other SSBs, and the protein is dynamically localized during DNA repair. Significance: The findings suggest a central role for DrSSB in DNA repair. The single-stranded DNA (ssDNA)-binding protein from the radiation-resistant bacterium Deinococcus radiodurans (DrSSB) functions as a homodimer in which each monomer contains two oligonucleotide-binding (OB) domains. This arrangement is exceedingly rare among bacterial SSBs, which typically form homotetramers of single-OB domain subunits. To better understand how this unusual structure influences the DNA binding and biological functions of DrSSB in D. radiodurans radiation resistance, we have examined the structure of DrSSB in complex with ssDNA and the DNA damage-dependent cellular dynamics of DrSSB. The x-ray crystal structure of the DrSSB-ssDNA complex shows that ssDNA binds to surfaces of DrSSB that are analogous to those mapped in homotetrameric SSBs, although there are distinct contacts in DrSSB that mediate species-specific ssDNA binding. Observations by electron microscopy reveal two salt-dependent ssDNA-binding modes for DrSSB that strongly resemble those of the homotetrameric Escherichia coli SSB, further supporting a shared overall DNA binding mechanism between the two classes of bacterial SSBs. In vivo, DrSSB levels are heavily induced following exposure to ionizing radiation. This accumulation is accompanied by dramatic time-dependent DrSSB cellular dynamics in which a single nucleoid-centric focus of DrSSB is observed within 1 h of irradiation but is dispersed by 3 h after irradiation. These kinetics parallel those of D. radiodurans postirradiation genome reconstitution, suggesting that DrSSB dynamics could play important organizational roles in DNA repair.


Molecular BioSystems | 2007

The tallysomycin biosynthetic gene cluster from streptoalloteichus hindustanus E465-94 ATCC 31158 unveiling new insights into the biosynthesis of the bleomycin family of antitumor antibiotics

Meifeng Tao; Liyan Wang; Evelyn Wendt-Pienkowski; Nicholas P. George; Ute Galm; Guodong Zhang; Jane M. Coughlin; Ben Shen

The tallysomycins (TLMs) belong to the bleomycin (BLM) family of antitumor antibiotics. The BLM biosynthetic gene cluster has been cloned and characterized previously from Streptomyces verticillus ATCC 15003, but engineering BLM biosynthesis for novel analogs has been hampered by the lack of a genetic system for S. verticillus. We now report the cloning and sequencing of the TLM biosynthetic gene cluster from Streptoalloteichus hindustanus E465-94 ATCC 31158 and the development of a genetic system for S. hindustanus, demonstrating the feasibility to manipulate TLM biosynthesis in S. hindustanus by gene inactivation and mutant complementation. Sequence analysis of the cloned 80.2 kb region revealed 40 open reading frames (ORFs), 30 of which were assigned to the TLM biosynthetic gene cluster. The TLM gene cluster consists of nonribosomal peptide synthetase (NRPS) genes encoding nine NRPS modules, a polyketide synthase (PKS) gene encoding one PKS module, genes encoding seven enzymes for deoxysugar biosynthesis and attachment, as well as genes encoding other biosynthesis, resistance, and regulatory proteins. The involvement of the cloned gene cluster in TLM biosynthesis was confirmed by inactivating the tlmE glycosyltransferase gene to generate a TLM non-producing mutant and by restoring TLM production to the DeltatlmE::ermE mutant strain upon expressing a functional copy of tlmE. The TLM gene cluster is highly homologous to the BLM cluster, with 25 of the 30 ORFs identified within the two clusters exhibiting striking similarities. The structural similarities and differences between TLM and BLM were reflected remarkably well by the genes and their organization in their respective biosynthetic gene clusters.


Nucleic Acids Research | 2011

Mechanism of Exonuclease I stimulation by the single-stranded DNA-binding protein

Duo Lu; Angela R. Myers; Nicholas P. George; James L. Keck

Bacterial single-stranded (ss) DNA-binding proteins (SSBs) bind and protect ssDNA intermediates formed during cellular DNA replication, recombination and repair reactions. SSBs also form complexes with an array of genome maintenance enzymes via their conserved C-terminal tail (SSB-Ct) elements. In many cases, complex formation with SSB stimulates the biochemical activities of its protein partners. Here, we investigate the mechanism by which Escherichia coli SSB stimulates hydrolysis of ssDNA by Exonuclease I (ExoI). Steady-state kinetic experiments show that SSB stimulates ExoI activity through effects on both apparent kcat and Km. SSB variant proteins with altered SSB-Ct sequences either stimulate more modestly or inhibit ExoI hydrolysis of ssDNA due to increases in the apparent Michaelis constant, highlighting a role for protein complex formation in ExoI substrate binding. Consistent with a model in which SSB stabilizes ExoI substrate binding and melts secondary structures that could impede ExoI processivity, the specific activity of a fusion protein in which ExoI is tethered to SSB is nearly equivalent to that of SSB-stimulated ExoI. Taken together, these studies delineate stimulatory roles for SSB in which protein interactions and ssDNA binding are both important for maximal activity of its protein partners.


Journal of Biological Chemistry | 2011

Structure and Biochemical Activities of Escherichia coli MgsA

Asher N. Page; Nicholas P. George; Aimee H. Marceau; Michael M. Cox; James L. Keck

Bacterial “maintenance of genome stability protein A” (MgsA) and related eukaryotic enzymes play important roles in cellular responses to stalled DNA replication processes. Sequence information identifies MgsA enzymes as members of the clamp loader clade of AAA+ proteins, but structural information defining the family has been limited. Here, the x-ray crystal structure of Escherichia coli MgsA is described, revealing a homotetrameric arrangement for the protein that distinguishes it from other clamp loader clade AAA+ proteins. Each MgsA protomer is composed of three elements as follows: ATP-binding and helical lid domains (conserved among AAA+ proteins) and a tetramerization domain. Although the tetramerization domains bury the greatest amount of surface area in the MgsA oligomer, each of the domains participates in oligomerization to form a highly intertwined quaternary structure. Phosphate is bound at each AAA+ ATP-binding site, but the active sites do not appear to be in a catalytically competent conformation due to displacement of Arg finger residues. E. coli MgsA is also shown to form a complex with the single-stranded DNA-binding protein through co-purification and biochemical studies. MgsA DNA-dependent ATPase activity is inhibited by single-stranded DNA-binding protein. Together, these structural and biochemical observations provide insights into the mechanisms of MgsA family AAA+ proteins.


Nucleic Acids Research | 2010

The crystal structure of Neisseria gonorrhoeae PriB reveals mechanistic differences among bacterial DNA replication restart pathways

Jinlan Dong; Nicholas P. George; Katrina L. Duckett; Madeleine A. Palacios DeBeer; Matthew Lopper

Reactivation of repaired DNA replication forks is essential for complete duplication of bacterial genomes. However, not all bacteria encode homologs of the well-studied Escherichia coli DNA replication restart primosome proteins, suggesting that there might be distinct mechanistic differences among DNA replication restart pathways in diverse bacteria. Since reactivation of repaired DNA replication forks requires coordinated DNA and protein binding by DNA replication restart primosome proteins, we determined the crystal structure of Neisseria gonorrhoeae PriB at 2.7 Å resolution and investigated its ability to physically interact with DNA and PriA helicase. Comparison of the crystal structures of PriB from N. gonorrhoeae and E. coli reveals a well-conserved homodimeric structure consisting of two oligosaccharide/oligonucleotide-binding (OB) folds. In spite of their overall structural similarity, there is significant species variation in the type and distribution of surface amino acid residues. This correlates with striking differences in the affinity with which each PriB homolog binds single-stranded DNA and PriA helicase. These results provide evidence that mechanisms of DNA replication restart are not identical across diverse species and that these pathways have likely become specialized to meet the needs of individual organisms.


Nature | 2009

Molecular biology: Slip sliding on DNA.

Nicholas P. George; James L. Keck

Dedicated binding proteins stabilize single-stranded DNA, protecting it from breakage and distortion. Once thought to form inert complexes with DNA, such proteins are now shown to be remarkably mobile.


Journal of Biological Chemistry | 2010

X-ray Crystal Structure of the Bacterial Conjugation Factor PsiB, a Negative Regulator of RecA

Vessela Petrova; Kenneth A. Satyshur; Nicholas P. George; Darrell R. McCaslin; Michael M. Cox; James L. Keck

During bacterial conjugation, genetic material from one cell is transferred to another as single-stranded DNA. The introduction of single-stranded DNA into the recipient cell would ordinarily trigger a potentially deleterious transcriptional response called SOS, which is initiated by RecA protein filaments formed on the DNA. During F plasmid conjugation, however, the SOS response is suppressed by PsiB, an F-plasmid-encoded protein that binds and sequesters free RecA to prevent filament formation. Among the many characterized RecA modulator proteins, PsiB is unique in using sequestration as an inhibitory mechanism. We describe the crystal structure of PsiB from the Escherichia coli F plasmid. The stucture of PsiB is surprisingly similar to CapZ, a eukaryotic actin filament capping protein. Structure-directed neutralization of electronegative surfaces on PsiB abrogates RecA inhibition whereas neutralization of an electropositive surface element enhances PsiB inhibition of RecA. Together, these studies provide a first molecular view of PsiB and highlight its use as a reagent in studies of RecA activity.

Collaboration


Dive into the Nicholas P. George's collaboration.

Top Co-Authors

Avatar

James L. Keck

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Aimee H. Marceau

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ben Shen

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Evelyn Wendt-Pienkowski

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jane M. Coughlin

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Liyan Wang

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Michael M. Cox

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Tae-Jin Oh

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ute Galm

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Angela R. Myers

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge