SCIENCE CHINA Life Sciences

SPECIAL TOPIC May 2012 Vol.55 No.5: 386–403 • REVIEW • doi: 10.1007/s11427-012-4324-9

Rapid progress of DNA replication studies in , the third domain of life

ISHINO Yoshizumi* & ISHINO Sonoko

Department of Bioscience and , Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 812-8581, Japan

Received March 27, 2012; accepted April 20, 2012

Archaea, the third domain of life, are interesting organisms to study from the aspects of molecular and evolutionary biology. Archaeal cells have a unicellular ultrastructure without a nucleus, resembling bacterial cells, but the proteins involved in ge- netic information processing pathways, including DNA replication, transcription, and translation, share strong similarities with those of Eukaryota. Therefore, archaea provide useful model systems to understand the more complex mechanisms of genetic information processing in eukaryotic cells. Moreover, the hyperthermophilic archaea provide very stable proteins, which are especially useful for the isolation of replisomal multicomplexes, to analyze their structures and functions. This review focuses on the history, current status, and future directions of archaeal DNA replication studies.

Archaea, DNA replication, DNA polymerase, replication fork, replisome, hyperthermophile

Citation: Ishino Y, Ishino S. Rapid progress of DNA replication studies in Archaea, the third domain of life. Sci China Life Sci, 2012, 55: 386–403, doi: 10.1007/ s11427-012-4324-9

The accurate duplication and transmission of genetic infor- Eukaryota evolved from a common ancestor, which was mation is essential and crucially important for living organ- different from that of Bacteria [5]. In addition, based on isms. The molecular mechanism of DNA replication has predictions from the genome size and the number of genes, been one of the central themes of molecular biology since the archaeal replication machinery is a simplified form of the discovery of the double helix DNA structure in 1953 [1]. that in eukaryotes, and is a good model to elucidate the Since then, molecular biologists have been making contin- functions of each component of the complex. On the other uous efforts to elucidate the precise mechanism of DNA hand, the circular genome structure is conserved in Bacteria replication. We now know the protein factors involved and and Archaea. These features have encouraged studies of each of their functions in the basic mechanism; from origin DNA replication in Archaea, in the hopes of gaining fun- of replication recognition to replication fork progression in damental insights into this process and its machinery from a semi-conservative replication manner, using continuous an evolutional perspective. Active research on archaeal and discontinuous strand syntheses. Archaeal homologs of DNA replication started in about 1990, as compared with many eukaryotic replication proteins (including ORC, Cdc6, bacterial studies before 1960 and eukaryotic studies before GINS, MCM, RPA, primase, DNA polymerase, PCNA, RFC, 1980. However, the progress in archaeal research has been FEN1, and DNA ligase) have been identified (Figure 1) and rapid, especially with the available total genome sequences, biochemically characterized [24]. Their similarities indi- and now the depth of knowledge of archaeal DNA replica- cate that the DNA replication machineries of Archaea and tion has almost caught up with that of the bacterial and eu-

karyotic research fields. In this review, we will summarize the research on the archaeal proteins involved in the DNA *Corresponding author (email: [email protected])

© The Author(s) 2012. This article is published with open access at Springerlink.com life.scichina.com www.springer.com/scp Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 387

Figure 1 DNA replication proteins and their functions in the three domains of life. Most of the archaeal proteins are designated as homologs of the proteins identified from eukaryotic DNA replication studies. replication process. microarray analysis, and showed that highly purified Cdc6/ Orc1 binds to the ORB and miniORB in the oriC region of P. furiosus in vitro [11]. 1 DNA replication-related proteins The crystal structure of the Cdc6/Orc1 protein from Py- 1.1 Origin of replication (oriC) and origin recognition robaculum aerophilum [12] and one of the two Cdc6/Orc1 proteins proteins, ORC2 (the authors referred to the two homologs in this organism as ORC1 and ORC2), from Aeropyrum pernix We identified the first archaeal gene encoding an amino [13] were determined. These Cdc6/Orc1 proteins consist of acid sequence similar to those of eukaryotic Cdc6 and Orc1 three structural domains. Domains I and II fold in a manner adjacent to the gene encoding DNA polymerase II (now similar to the AAA+ family proteins. Domain III has a called PolD, as described below) in the Pyrococcus furiosus winged helix (WH) fold, which is present in a number of genome [6]. Subsequently, we identified the origin of repli- DNA binding proteins. There are four ORBs arranged in cation (oriC) just upstream of the gene encoding the Cdc6 pairs on both sides of the DUE in the oriC region of A. and Orc1-like sequences in the Pyrococcus genome [7]. We pernix, and ORC1 binds to each ORB as a dimer. A mecha- named the gene product Cdc6/Orc1, because of its roughly nism was proposed in which after all four ORBs have bound equal homology to regions of eukaryotic Orc1 and Cdc6, ORC1 proteins, a higher-order assembly of the origin initi- after confirming that the protein actually bound to the oriC ates unwinding of the DUE, with alterations in both topol- region on the chromosomal DNA [7]. The archaeal genome ogy and superhelicity [14]. Furthermore, two reports de- sequences had conserved 13 bp repeats characteristic of scribed the crystal structures of S. solfataricus Cdc6-1 and oriC, as predicted by bioinformatics [8]. In Pyrococcus ge- Cdc6-3 (two of the three Cdc6/Orc1 proteins in this organ- nomes, two of the repeats were longer than the conserved ism) forming a heterodimer bound to ori2 DNA (one of the 13 bp and surrounded a putative DNA unwinding element three origins in this organism) [15], and that A. pernix (DUE) with an AT-rich sequence (Figure 2) [9]. The longer ORC1 bound to an origin sequence [16]. These studies re- repeated sequence was designated as an Origin Recognition vealed that both the N-terminal AAA+ ATPase domain Box (ORB), and was recognized by Cdc6/Orc1 in a Sulfol- (domain I+II) and C-terminal winged-helix (WH) domain obus solfataricus study [10]. The 13 base repeat was called (domain III) contribute to origin DNA binding. In addition, a miniORB, being a minimal version of an ORB. We found the structural information not only defined the polarity of that P. furiosus Cdc6/Orc1 binds to the oriC region in the initiator assembly on the origin, but also indicated the in- genome with extreme specificity, using a whole genome duction of substantial distortion into the DNA strands. This 388 Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5

Figure 2 Detailed structure of the oriC region in the Pyrococcus genome. oriC, located in the upstream region of the cdc6/orc1 gene, is schematically drawn with the exact nucleotide sequences around the first unwinding site, identified by P1 nuclease assays, and the initiation site for primer synthesis, iden- tified by a RIP assay. The consensus sequences of ORB1 and ORB2 (origin recognition box) are indicated in red, and the 12 bp P1-sensitive site is indicat- ed in blue. The region coding for Cdc6/Orc1 is underlined.

Table 1 Distribution of DNA polymerases from seven families in the three domains of lifea)

Family Bacteria Archaea Eukaryote Euryarchaeota Korarchaeota Aigarchaeota Thaumarchaeota

A Pol I Pol θ Pol γ**

Pol BI Pol BI B Pol II Pol I(B) Pol BII Pol BII Pol α, Pol δ Pol ε, Pol ζ Pol BII Pol BII C Pol III

D Pol D Pol D Pol D Pol D

E Pol E*

X Pol β, Pol λ Pol μ, Pol σ

Pol IV Y Pol η, Pol ι Pol κ Pol V a) *, plasmid-coded; **, mitochondrial. distortion probably triggers the unwinding of the duplex not fully understood in archaea, it should minimally include DNA to begin replication. Furthermore, structural data pro- MCM, GINS, primase, PCNA, DNA polymerase, and RPA, vided the detailed interaction mode between the initiator as described below. The assembly of these factors must be protein and the oriC DNA. Mutational analyses of the M. within the vicinity of the single-stranded region. thermautotrophicus Cdc6-1 protein also revealed the essen- The following studies revealed that the ATPase activity tial interaction between an arginine residue conserved in the of the Cdc6/Orc1 protein was completely suppressed by archaeal Cdc6/Orc1 and an invariant guanine in the ORB binding to DNA containing the origin recognition box sequence [17]. (ORB). Limited proteolysis and DNase I-footprint experi- Following the discovery of P. furiosus Cdc6/Orc1 in ments suggested that the Cdc6/Orc1 protein changes its 1995 [6], difficulties in preparing highly purified protein in conformation on the ORB sequence in the presence of ATP. a soluble fraction made it difficult to characterize the pro- The reason for this conformational change is unknown, but tein. In our recent report using protein prepared in a dena- it may have an important function in the initiation process turation-renaturation procedure [18], we found that the P. [19]. In addition, results from an in vitro recruitment assay furiosus Cdc6/Orc1 protein alone can unwind duplex DNA indicated that Mcm (minichromosome maintenance), the at a specific site in the oriC region in vitro, in an ATP- replicative DNA helicase (described below), is recruited independent manner. The local unwinding site we identified into the oriC region in a Cdc6/Orc1-dependent, but not is approximately 670 bp downstream from the transition site ATP-dependent, manner [19]. However, this recruitment is between leading and lagging syntheses (Figure 2), as deter- not sufficient for the unwinding function of Mcm, which mined by an in vivo replication initiation point (RIP) assay indicates another protein remains to be identified for the [9]. The replication machinery at the unwound site is still to functional loading of this helicase to start the progression of be established. Although the details of the machinery are the replication fork DNA. Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 389

Table 2 The archaeal DNA ligases characterized to date and their cofac- tor utilization

Species Cofactor Reference Acidithiobacillus ferrooxidans ATP [165]

Aeropyrum pernix ATP ADP [166] Ferroplasma acidarmanus ATP [197] + Ferroplasma acidophilum ATP NAD [165] Methanothermobacterium ATP [198] thermoautotrophicum + Picrophilus torridus ATP NAD [165] Pyrococcus horikoshii ATP [169]

Pyrococcus furiosus ATP [170]

Staphylothermus marinus ATP ADP [167] Sulfophobococcus zilligii ATP ADP GTP [168] Sulfolobus acidocaldarius ATP [165]

Sulfolobus shibatae ATP [199] + Thermococcus fumicolans ATP NAD [163] + Thermococcus kodakarensis ATP NAD [162] + Thermococcus sp. ATP NAD [164] Figure 3 Replicative helicase complexes in eukaryotes and archaea. The CMG complex is the core of the helicase for the template DNA unwinding Thermococcus sp. 1519 ATP [200] + reaction in eukaryotes. The archaeal genomes contain the homologs of the Thermoplasma acidophilum ATP NAD [165] Mcm and Gins proteins, but a Cdc45 homolog has not been identified. Recent research suggests that a RecJ-like exonuclease, with weak sequence Analyses of the multiple origins (more than two oriCs in homology to that of Cdc45, may work as a helicase complex with MCM and GINS, although the exact role of its 5′–3′ exonuclease remains un- a single chromosomal DNA), identified on several archaeal known. genomes, have primarily been performed in Sulfolobus strains [10,2024]. This is also an interesting subject in the research field of archaeal DNA replication. Questions re- munoprecipitation (ChIP) method in P. furiosus, especially main regarding how the initiations of replication from mul- in the exponentially growing phase [7,11]. We have also tiple origins are regulated, and how the replication forks been studying the MCM helicase of P. furiosus in vitro, and progress after the collision of two forks from opposite di- could not detect significant helicase activity from the puri- rections. fied protein. However, the DNA helicase activity was clearly stimulated by the addition of GINS (the Gins23- Gins51 complex), which is the homolog of the eukaryotic 1.2 MCM helicase GINS complex (described below in more detail). This Another important protein involved in the initiation of DNA result suggests that GINS works as an accessory factor to replication is the helicase, which unwinds duplex DNA for form a replication fork progression complex (RPC) in P. replication fork progression. The MCM protein complex, furiosus [31]. consisting of six subunits (Mcm2–7), is the replicative hel- The interactions between the Cdc6/Orc1 and Mcm pro- icase “core” in eukaryotic cells [25]. The MCM complex teins have been reported in several archaeal organisms, in- further forms a higher complex with Cdc45 and GINS cluding P. furiosus, as described above. It was found that (Sld5-Psf1-Psf2-Psf3), and this complex, called the CMG the two Cdc6/Orc1 homologs, Cdc6-1 and Cdc6-2, both complex, is now believed to be the functional helicase in inhibit the helicase activity of MCM (Mcm complex) in eukaryotic cells (Figure 3) [26]. Methanothermobacter thermoautotrophicus [33,34]. The Most archaeal genomes appear to encode at least one Cdc6-1 protein also inhibits the helicase activity of MCM in Mcm homolog, and the helicase activities of these proteins S. solfataricus [35]. In contrast, the Cdc6-2 protein stimu- from several archaeal organisms have been confirmed in lates the helicase activity of Mcm in Thermoplasma aci- vitro [2731]. In contrast to the eukaryotic MCM complex, dophilum [36]. these archaeal MCMs, consisting of a hexamer or double One interesting feature of archaea is that several hexamer, have distinct DNA helicase activity by themselves have multiple chromosomal genes encoding Mcm homologs. in vitro. Therefore, these MCMs may function on their own Recent comprehensive genomic analyses revealed that 13 as the replicative helicase in vivo. The structure-function archaeal species have more than one mcm gene. Genetic relationships of the archaeal Mcms have been aggressively context analysis showed that many of the mcm genes in ar- studied [32]. We reported that Mcm preferentially binds to chaeal genomes reside within mobile elements, originating the origin in vivo, as determined using a chromatin im- from viruses and plasmids [37]. For example, Thermococ- 390 Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 cus kodakarensis has three mcm genes [38]. Two of Mcm1 co-precipitated with Mcm3 and GINS, although they the three genes are located in regions where genetic ele- did not form a heterohexameric complex [42]. This sug- ments have presumably been integrated into the genome. gested that Mcm1 is involved in the replisome or repair- However, these genes are apparently stably inherited, and some, and shares some function in T. kodakarensis cells. their gene products may perform some important functions Further phenotypic analyses investigating the sensitivities of in the DNA metabolism of T. kodakarensis. The establish- the ∆mcm1 and ∆mcm2 mutant strains to DNA damage ment of a genetic manipulation system, the first for a caused by various mutagens, as reported for other DNA hyperthermophilic euryarchaeon, in T. kodakarensis [39,40], repair-related genes in T. kodakarensis [44], may help elu- is an advantage for investigating the function of the Mcm cidate the functions of these Mcm proteins. proteins. Gene disruption experiments were recently per- In the case of the Methanococcus maripaludis S2 strain, formed for each mcm gene by two groups, including ours which harbors four mcm genes (three of which seem to be [41,42]. These experiments revealed that mcm3 cannot be derived from phage), a shotgun proteomics study detected disrupted, in contrast to the other two mcm genes which peptides originating from three out of the four mcm gene were easily knocked out. This fact, as well as its relative products [45]. Furthermore, the four gene products abundance in the cells, indicates that Mcm3 is the main hel- co-expressed in E. coli were co-purified in the same fraction icase core protein in the normal DNA replication process in [46]. These results suggest that multiple Mcm proteins are T. kodakarensis. The stability of the hexameric structure in functional in archaeal cells. solution, found only for Mcm3, also supports the proposal A further important issue is how Mcm is recruited onto that the Mcm3 protein is the predominant helicase in DNA the unwound region of oriC and whether Cdc6/Orc1 is the replication. sole recruiter. Additional critical recruiting factors may ex- Therefore, the function of the other two Mcm proteins ist in archaeal cells. The relationship between the ATPase remains unclear. The research group of Reeve and Kelman activity of Cdc6/Orc1 and the Mcm loading of the oriC re- recently reported an investigation of the network between gion is also a key question to be solved. Further studies are DNA replication-related proteins in T. kodakarensis [43]. required to understand the ultimate principles of DNA rep- They introduced a His-tag sequence into 19 different genes lication initiation. in the T. kodakarensis genome that encode DNA replica- tion-related proteins. Protein complexes were purified from 1.3 GINS exponentially growing cells of each strain by His-tag affin- ity chromatography (Ni2+-chelating column), and the frac- The eukaryotic GINS complex consists of four different tions were subjected to mass spectrometry (MS) analysis to proteins, Sld5, Psf1, Psf2, and Psf3 (GINS is an acronym detect the proteins included in each complex. They detected for the Japanese go-ichi-ni-san, meaning 5-1-2-3, after these the complex from a strain expressing the His-tagged Mcm2 four subunits), originally identified in Saccharomyces cere- protein, indicating that the His-tagged Mcm2 could be ef- visiae as essential protein factors for the initiation of DNA fectively produced in the strain. Furthermore, they showed replication [47]. The amino acid sequences of the four sub- that the recombinant His-tagged Mcm2 has ATPase and units in the GINS complex share some conservation, sug- helicase activities in vitro. However, our western blot anal- gesting that they are ancestral paralogs [48]. Finely detailed ysis could not detect TkoMcm2 in the extract from expo- bioinformatics analyses identified the homologs of GINS nentially growing T. kodakarensis cells. Our subsequent subunits in archaeal genomes; however, most of the archaea reverse-transcriptase PCR (RT-PCR) experiment detected have only one gene encoding this family protein, and more mcm2 gene transcript in the cells (Ishino et al., unpublished interestingly, the crenarchaea and euryarchaea (the two ma- result). Therefore, the mcm2 gene appears to be expressed jor subdomains of archaea) characteristically have sequenc- under normal growth conditions. It is also possible that the es more similar to Psf2 and Psf3, and Sld5 and Psf1, respec- expression of mcm2 is constitutive, but the Mcm2 protein is tively [31,48]. A GINS homolog, designated Gins23, was degraded rapidly. Further experiments to measure the effi- detected in S. solfataricus in a yeast two-hybrid screen for ciency of mcm2 gene transcription by quantitative PCR, as interaction partners of MCM. Another subunit, designated well as to assess the stability of the Mcm2 protein in the cell Gins15, was identified by the MS analysis of an immu- extract, are needed. Our experiments demonstrated robust noaffinity-purified native GINS complex from a S. solfata- DNA unwinding activities in vitro from two of the three ricus cell extract, using an anti-GINS23 antibody [49]. The Mcm proteins. The helicase activity of Mcm1 is strong in S. solfataricus GINS complex, composed of Gins23 and vitro, and a distinct amount of Mcm1 protein is present in T. Gins15, forms a tetrameric structure with a 2:2 molar ratio kodakarensis cells. Moreover, Mcm1 functionally interacts [49]. We also reported the GINS complex from P. furiosus, with the GINS complex from T. kodakarensis [42]. These a complex of Gins23 and Gins51 with a 2:2 ratio, as the first facts strongly suggest that Mcm1 does participate in some euryarchaeal GINS (we prefer to call the GINS subunit pro- aspect of DNA transactions, and may be substituted with teins Gins23 and Gins51 (instead of Gins15), consistent Mcm3. Our immunoprecipitation experiments showed that with the original nomenclature of GINS from 5-1-2-3 [31]. Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 391

The Mcm2–7 hexamer was co-purified in complex with lum was constructed. A long disordered region inserted be- Cdc45 and GINS from Drosophila melanogaster embryo tween the A and B domains allows the differential position- extracts and S. cerevisiae lysates. The Cdc45-MCM2–7- ing of the C-terminal domains and, as a consequence, ex- GINS (CMG) complex (Figure 3), as described above, was clusively leads to the formation of an asymmetric homo- also associated with the replication fork in Xenopus laevis tetramer, rather than a symmetrical assembly [52]. egg extracts, and a large molecular machine, containing The Cdc45 protein is ubiquitously distributed from yeast Cdc45, GINS, and MCM2–7, was proposed as the un- to humans, however its exact role in the CMG complex has windosome to separate the DNA strands at the replication not been elucidated. No archaeal homolog of Cdc45 has fork [50]. These results indicate that GINS is a critical fac- been identified. A detailed analysis of the primary structure tor for not only the initiation process, but also the elonga- of Cdc45 revealed that eukaryotic Cdc45 and prokaryotic tion process in eukaryotic DNA replication. S. solfataricus RecJ share a common ancestry [56]. A homolog of the GINS interacts with MCM and primase, although no stimu- DNA binding domain of RecJ was co-purified with GINS lation or inhibition of either the helicase or primase activity from S. solfataricus [49]. We detected stimulation of the was observed by the interaction with GINS in vitro [49]. 5′–3′ exonuclease activity of the RecJ homologs from P. Conversely, the DNA helicase activity of P. furiosus MCM furiosus and T. kodakarensis by the cognate GINSs (Ishino is clearly stimulated by the addition of the P. furiosus GINS et al., unpublished result). A recent report found that the complex, as described above [31]. RecJ homolog from T. kodakarensis forms a stable complex T. acidophilum, like many euryarchaea, has only one with the T. kodakensis GINS, and that the 5′–3′ exonuclease gene sequence homologous to Gins51 in its genome. Gel activity is enhanced in vitro (therefore, the RecJ homolog filtration and electron microscopy analyses revealed that T. was designated GAN, from GINS-associated nuclease) [57]. acidophilum Gins51 forms a homotetramer. A physical in- Another related report found that the human Cdc45 protein teraction between T. acidophilum Gins51 and Mcm was could bind single-stranded, but not double-stranded, DNA. detected by surface plasmon resonance analysis (SPR). The structure obtained by small angle X-ray scattering TaGins51 did not affect the helicase activity of its cognate analysis was consistent with a model compatible with the MCM when an equal ratio of each molecule was tested in vitro [51]. These results suggest that another factor is re- crystallographic structure of the RecJ family members [58]. quired to form a stable helicase complex with MCM and These interesting findings will promote further research on GINS at the replication fork in T. acidophilum. However, an the structures and functions of the higher-order unwindo- excess amount of TaGINS clearly stimulated the helicase some in archaeal cells (Figure 3). activity (Ogino et al., unpublished result). In the case of T. kodakarensis, the ATPase and helicase activities of MCM1 1.4 Primase and MCM3 were clearly stimulated by T. kodakarensis GINS in vitro. It is noteworthy that the helicase activity of To initiate DNA strand synthesis, a primase is required for MCM1 was stimulated more than that of MCM3. Physical the synthesis of a short oligonucleotide primer. DnaG and interactions between the T. kodakarensis Gins and Mcm p48-p58 are well-known primases in Bacteria and Eukaryo- proteins were also detected [42]. These reports suggested ta, respectively. The p48-p58 primase is further complexed that the MCM-GINS complex is a common part of the rep- with p180 and p70, in what is called the DNA polymerase licative helicase in Archaea (Figure 3). α-primase complex. The catalytic subunits of the eukaryotic Recently, we determined the crystal structure of the T. and archaeal primases, which synthesize a short primer on kodakarensis GINS tetramer, comprising Gins51 and the template DNA, share some sequence homology with Gins23, and compared it with the reported human GINS those of the family X DNA polymerases [59]. The first ar- structures [52]. Each subunit of human GINS consists of an chaeal primase was identified in Methanococcus jannaschii, α-helical main domain and a β-stranded small domain, and as an ORF with a sequence similar to that of the catalytic assembles into the heterotetramer, which exhibits a unique subunit, p48, of the eukaryotic primase. The gene product trapezoidal shape [5355]. The four subunits share a similar exhibited the ability to synthesize oligonucleotides on the fold, and they are classified into two groups: Sld5 and Psf1 template DNA [60]. We characterized the p48-like protein possess the α-helical (A) domain at the N-terminus and the (p41) from P. furiosus. Unexpectedly, the archaeal p41 pro- β-stranded domain (B) at the C-terminus (AB-type), while tein did not catalyze the synthesis of short RNA by itself, Psf2 and Psf3 are the permuted version (BA-type). The but preferentially used deoxynucleotides to synthesize DNA backbone structure of each subunit and the tetrameric as- strands up to several kilobases in length [61]. We found that sembly of T. kodakarensis GINS are similar to those of hu- the gene neighboring the p41 gene encodes a protein with man GINS. However, the location of the C-terminal B do- some similarity to the p58 subunit of eukaryotic primase. main of Gins51 is remarkably different between the two This gene product, named p46, forms a stable complex with GINS structures [52]. Based on crystal structures, a homol- p41, and the complex can synthesize a short RNA primer in ogy model of the homotetrameric GINS from T. acidophi- vitro [62], consistent with the fact that the short RNA pri- 392 Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 mer was identified in Pyrococcus cells [9]. However, this 1.5 Single-stranded DNA binding protein primase complex also synthesizes DNA strands several The single-stranded DNA binding protein, called SSB in hundred nucleotide in length in vitro [62]. Bacteria and RPA (replication protein A) in Archaea and Further reports characterizing the homologs from S. sol- Eukaryota, is important for protecting the unwound sin- fataricus [6365], P. horikoshii [6668], and P. abyssi [69] gle-stranded DNA from nuclease attack, chemical modifica- revealed that these primases have similar properties in vitro. tion, and other disruptions during the DNA replication pro- Notably, the small subunit, p41 (PriS), is the catalytic subu- cess. Although there is little amino acid sequence similarity nit, while the large subunit, p46 (PriL), modulates the activ- between SSB and RPA, the three-dimensional structures ity in the heterodimeric archaeal primases. The crystal revealed that these proteins have a structurally similar do- structure of the N-terminal domain of PriL complexed with main containing a common fold, called the oligonucleo- PriS of S. solfataricus primase revealed that PriL does not tide/oligosaccharide-binding (OB) fold [75]. The common directly contact the active site of PriS, and therefore, the structure in SSB and RPA suggests that the mechanism of large subunit may interact with the synthesized primer to single-stranded nucleic acid binding is conserved in living adjust its length to 7–14 mer. The structure of the catalytic organisms. In Bacteria, E. coli SSB is a homotetramer of a center, containing a triple aspartic acid motif, is similar to 20 kD peptide with one OB-fold, and the SSBs from Dein- those of the family X DNA polymerases, as predicted from ococcus radiodurans and Thermus aquaticus consist of a the amino acid sequence similarities. The 3′-terminal nucle- homodimer of the peptide containing two OB-folds. In con- otidyl transferase activity, detected in the S. solfataricus trast, the eukaryotic RPA is a stable heterotrimer, composed primase [63,65], and the gap-filling and strand-displacement of 70, 32, and 14 kD proteins. RPA70 contains two tandem activities of the P. abyssi primase [69] also support the repeats of an OB-fold, which are responsible for the major structural similarity between PriS and the family X en- interaction with a single-stranded DNA in its central region. zymes. The N-terminal and C-terminal regions of RPA70 mediate A further unique activity, named template-dependent interactions with many cellular or viral proteins, in addition polymerization across discontinuous template (PADT), has to RPA32 [76,77]. The middle subunit, RPA32, contains an recently been reported for S. solfataricus PriSL [70]. Huang OB-fold in the central region [78–80]. The C-terminal re- and co-authors propose that the activity may be involved in gion of RPA32 interacts with other RPA subunits and vari- double-stranded break repair in Archaea. ous cellular proteins [76,77,81,82]. The smallest subunit, Archaeal genomes also encode genes resembling bacteri- RPA14, also contains an OB-fold [78]. The eukaryotic RPA al DnaG primase. We expressed the gene encoding function has been well studied in the in vitro SV40 DNA the DnaG homolog from P. furiosus, however the protein replication system. RPA interacts with the SV40 T-antigen did not show any primer synthesis activity in vitro, and thus and the DNA polymerase α-primase complex, forming an initiation complex at the origin of replication [83]. RPA also the archaeal DnaG-like protein does not appear to be a pri- stimulates Polα-primase activity and PCNA-dependent Polδ mase (Fujikane et al. unpublished result). The DnaG-like activity [84,85]. protein was shown to participate in RNA degradation as an The RPAs from M. jannaschii and M. thermoauto- exosome component [71,72]. However, a recent report re- trophicus were reported in 1998, as the first archaeal sin- vealed that a DnaG homolog from S. solfataricus synthe- gle-stranded DNA binding proteins [8688]. These proteins sizes primers 13 nucleotides in length, which is the first share amino acid sequence similarity with eukaryotic evidence for the function of the DnaG homolog in Archaea RPA70, and contain four or five repeated domains with the [73]. It would be interesting to investigate if the two pri- putative OB-fold and one putative zinc-finger motif. M. mases, PriSL and DnaG, share the primer synthesis func- jannaschii RPA exists as a monomer in solution, and has tions for leading and lagging strand replication in archaeal single-strand DNA binding activity. We characterized the cells, as was suggested by the authors [73]. An interesting RPA from P. furiosus [89]. Similar to the eukaryotic RPA, hypothesis for the evolution of PriSL and DnaG from the P. furiosus RPA forms a complex consisting of three dis- last universal common ancestor (LUCA) has also been pro- tinct subunits. This RPA highly stimulated the RadA- posed by Hu et al. [70]. promoted strand-exchange reaction in vitro [89]. The Sulfolobus PriSL protein was shown to interact with While the euryarchaeal organisms have a eukaryotic-type MCM through Gins23 [49]. This interaction probably en- RPA homolog, the crenarchaeal SSB proteins appear to be sures the coupling of DNA unwinding and priming during much more similar to the bacterial proteins, with a single the replication fork progression [49]. Furthermore, the di- OB fold and a flexible C-terminal tail. The crystal structure rect interaction between PriSL and the clamp loader RFC of the SSB protein from S. solfataricus was determined to (described below) in S. solfataricus may regulate the primer 1.26 Å [90]. The structure of the OB-fold domain is more synthesis and its transfer to DNA polymerase in archaeal similar to that of the eukaryotic RPAs, although it was ex- cells [74]. pected to resemble the E. coli SSB, supporting the close Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 393 relationship between archaea and eukaryotes [90]. archaeal cells is still not known, although the distribution of The three subunits, RPA1, RPA2, and RPA3, from the DNA polymerases is better understood (Table 1) [102]. Methanosarcina acetivorans were also characterized [91]. We cloned the gene encoding a family B DNA polymer- Unlike the multiple RPA proteins found in other archaea ase from P. furiosus, the first known entire pol gene in ar- and eukaryotes, each of the M. acetivorans RPAs can act chaea [103], while the family B DNA polymerases from as a distinct single-stranded DNA-binding protein. The Pyrodictium occultum [99] and A. pernix [100] were cloned architecture of the M. acetivorans RPA also differs from in the 1990s. The most interesting features discovered at the that found in other archaea. Each of the three RPA proteins, inception of this research area were that archaea possess the as well as their combinations, clearly stimulates the primer eukaryotic Polα-like (Family B) DNA polymerases [103], extension activity of M. acetivorans DNA polymerase BI in and some archaea (crenarchaea) have multiple Polα-like vitro, as shown previously for bacterial SSB and eukaryotic DNA polymerases, as in eukaryotic cells [99,100]. Lastly, a RPA [91]. Structural analyses of the architectures of SSB novel DNA polymerase (Family D) without any sequence and RPA suggested that they are composed of different homology was also discovered [6]. The early stages of the combinations of the OB-fold. Bacterial and eukaryotic research on the thermostable DNA polymerases have been organisms contain one type of SSB or RPA, respectively. summarized in a previous review article [104]. In contrast, archaeal organisms have various RPAs, com- The first family D DNA polymerase was identified from posed of different organizations of OB-folds. M. P. furiosus, by screening for DNA polymerase activity in acetivorans RPA1, 2, and 3 have four, two, and two OB- the cell extract [105]. Three independent fractions showing folds, respectively. The hypothesis that homologous recom- deoxynucleotide incorporation activity were obtained from bination might play an important role in generating the di- anion-exchange chromatography. DNA polymerase activity, versity of OB-folds in archaea was proposed, based on ex- which was not representative of a family B-like enzyme periments characterizing the engineered RPAs with various because of its resistance to aphidicolin (α-like DNA poly- OB-folds [92]. merase-specific DNA polymerase), was found in one frac- tion. The corresponding gene was cloned, revealing that this 1.6 DNA polymerase new DNA polymerase consisted of two proteins, named DP1 and DP2, and that the deduced amino acid sequences DNA polymerase catalyzes phosphodiester bond formation of these proteins were not conserved in the DNA polymer- between the terminal 3′-OH of the primer and the ase families [6]. This DNA polymerase, proposed as PolD α-phosphate of the incoming triphosphate. DNA polymer- from the new family (family D), has only been identified in ases have been classified by amino acid sequence similarity, the euryarchaeal organisms to date [94,101]. P. furiosus and seven families, A, B, C, D, E, X, and Y, are now widely PolD exhibits efficient strand extension activity and strong recognized (Table 1) [9397]. The fundamental ability of proofreading activity [6,106]. Other family D DNA poly- DNA polymerases to synthesize a deoxyribonucleotide merases were also characterized by several groups chain is conserved in relation to the structural conservation. [107–113]. However, the more specific properties, including processiv- A genetic study of Halobacterium sp. NRC-1 showed ity, synthesis accuracy, and substrate nucleotide selectivity, that both PolB and PolD are essential for viability [114]. An differ among the enzymes. Enzymes within the same family interesting issue is to elucidate whether PolB and PolD have similar basic properties. E. coli has five DNA poly- work together at the replication fork for the synthesis of the merases, and PolI, PolII, and PolIII belong to families A, B, leading and lagging strands, respectively. According to its and C, respectively. PolIV and PolV are classified in family biochemical properties, including the usage of an RNA Y, as the DNA polymerases for trans-lesion synthesis (TLS). primer and the presence of strand displacement activity, In eukaryotes, the replicative DNA polymerases, Polα, Polδ, PolD may catalyze lagging strand synthesis [102,112]. and Polε belong to family B, and the trans-lesion DNA The PolD genes were found only in Euryarchaeota, as polymerases, η, ι, and κ belong to family Y [98]. described above. However, recent environmental genomics Members of the Crenarchaeota have at least two family B and cultivation efforts revealed novel phyla in Archaea: DNA polymerases [99,100]. However, there is only one Thaumarchaeota, Korarchaeota, and Aigarchaeota, and their family B DNA polymerase in the Euryarchaeota. Instead, genome sequences showed that Thaumarchaeota and the euryarchaeotic genomes encode a family D DNA poly- Aigarchaeota harbor the genes encoding PolD and crenar- merase, as described below, which seems specific to these chaeal PolBII [115,116] , while Korarchaeota encodes PolBI, archaeal organisms, as it has never been found in any other PolBII and PolD [117]. In spite of accumulating infor- living organisms [94,101]. Several archaea have family mation about the distribution of DNA polymerases among Y-like DNA polymerases, although they have not been novel archaeal phyla (Table 1), only one gene for PolBII found in many other archaeal organisms. We will not de- from the psychrophilic marine thaumarchaeon, Cenar- scribe the trans-lesion synthesis by family Y DNA poly- chaeum symbiosum, has been analyzed [118]. Biochemical merase in this review. The role of each DNA polymerase in characterization of these gene products will contribute to 394 Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 research on the evolution of DNA polymerases in living DNA strand into the central hole. The γ-complex in Bacteria organisms. A hypothesis that the archaeal ancestor of eu- and replication factor C (RFC) in Archaea and Eukaryota karyotes encoded three DNA polymerases, two distinct are clamp loaders (Figure 4). We have been studying the family B polymerases and a family D polymerase, which all structures and functions of P. furiosus PCNA [130134] contributed to the evolution of the eukaryotic replication and RFC [135138] and found that these molecules are es- machinery, consisting of Polα, Polδ, and Polε, has been sential for DNA polymerase to perform processive DNA proposed [119]. synthesis. RFC loads PCNA onto the DNA strand in an One unique feature of the archaeal family B DNA poly- ATP-dependent manner. The mechanism of clamp loading merases is their specific interactions with uracil and hypo- is an interesting subject, and several research groups are xanthine, which stall the progression of the polymerases on actively investigating it [139] (Figure 4). We detected an DNA template strands [120]. This phenomenon, called intermediate PCNA-RFC-DNA complex, in which the “read-ahead recognition” of deaminated bases, was origi- PCNA ring is opened, by single particle analysis of electron nally proposed to be characteristic of hyperthermophilic microscopic images using P. furiosus proteins (Figure 4) archaea, to protect them against increased deamination at [140]. These results contributed to our understanding of the high temperature. However, the family B DNA polymerase molecular mechanism of the clamp loading process [140]. from the mesophilic archaeon, M. acetivorans, also recog- The crystal structure of the complex, including the nizes uracil and hypoxanthine as efficiently as the enzymes ATP-bound clamp loader, the ring-opened clamp, and the from hyperthermophiles [121]. The crystal structure of the template-primer DNA, using proteins from bacteriophage DNA polymerase revealed that read-ahead recognition oc- T4, has recently been published [141]. curs by an interaction with the deaminated bases in an Following clamp loading, DNA polymerase accesses the N-terminal binding pocket, existing specifically in the ar- PCNA, and the polymerase-clamp complex catalyzes pro- chaeal family B DNA polymerases [122]. cessive DNA synthesis. Therefore, structural and functional An interesting protein was identified in the plasmid studies of the DNA polymerase-PCNA interaction are the pRN1, isolated from a Sulfolobus strain [123]. This protein, next target to elucidate the overall mechanisms of replica- ORF904 (named RepA), has primase and DNA polymerase tion fork progression. Extensive studies of the PCNA- activities in the N-terminal domain, and helicase activity in interacting proteins revealed that the proteins contain a the C-terminal domain, and is likely to be essential for the small conserved sequence motif, called the PCNA interact- replication of pRN1 [124,125]. The amino acid sequence of the N-terminal domain lacks homology to any known DNA polymerase or primase, although the C-terminal domain contains a sequence conserved in the superfamily 3 helicas- es, and therefore, family E was proposed as a new DNA polymerase family. Similar proteins are encoded by various archaeal and bacterial plasmids, as well as by some bacterial viruses [126]. Recently, protein t2-12p, encoded in the plasmid pTN2 isolated from Thermococcus nautilus, was experimentally identified as a DNA polymerase from this family [127]. This enzyme is likely to be responsible for plasmid replication. Further investigation of this family of DNA polymerases will be interesting from an evolutionary perspective.

1.7 PCNA and RFC

The sliding clamp is the processivity factor that retains the DNA polymerase on the template DNA strand for continu- ous strand synthesis. The doughnut-shaped ring structure of clamp molecules is conserved among living organisms. The eukaryotic and archaeal proliferating cell nuclear antigens Figure 4 Mechanisms of clamp loading and processive DNA synthesis. The clamp loader (RFC) opens and recruits the clamp (PCNA) ring onto (PCNAs) form a homotrimeric ring structure [128,129] that the primer terminus of the DNA strand, in an ATP-dependent manner. The encircles the DNA strand and anchors the DNA polymerase clamp loader is then replaced by DNA polymerase, which can synthesize onto the DNA by binding it on one surface (front side) of the DNA strand processively without falling off. The crystal structure of P. the ring, as described below (Figure 4). To load the clamp furiosus PCNA (PDB code 1GE8) and the structure of the clamp loading complex (PCNA-RFC-DNA ternary complex), determined by EM single ring onto the DNA strand, a clamp loader is required to in- particle analysis, are shown on the left. Each subunit of the PCNA ring is teract with the clamp and open its ring to introduce the shown in a different color. Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 395 ing protein box (PIP box), which binds to a common site on switching mechanism between the editing and synthesis PCNA [142]. The PIP box consists of the sequence modes [147]. “Qxxhxxaa”, where “x” represents any amino acid, “h” In contrast to almost all organisms in Euryarchaeota, represents a hydrophobic residue (e.g., L, I or M), and “a” which have a single PCNA homolog forming a homotri- represents an aromatic residue (e.g., F, Y or W). Archaeal meric ring structure, the majority of Crenarchaeota have DNA polymerases have PIP box-like motifs in their se- multiple PCNA homologs, which are capable of forming quences [143]. However, only a few studies have experi- heterotrimeric rings for their functions [148,149]. It is espe- mentally investigated the function of the motifs. cially interesting that the three PCNAs, PCNA1, PCNA2, We determined the crystal structure of P. furiosus PolB and PCNA3, specifically bind PCNA binding proteins, in- complexed with a monomeric PCNA mutant, which allowed cluding DNA polymerases, DNA ligases, and FEN-1 endo- us to construct a convincing model of the polymerase- nuclease [149,150]. Detailed structural studies of the heter- PCNA ring interaction [144]. Our structural study revealed ologous PCNA from S. solfataricus revealed that the inter- that a novel interaction is formed between a stretched loop action modes between the subunits are conserved with those of PCNA and the thumb domain of PolB, in addition to the of the homotrimeric PCNAs [151,152]. authentic PIP box. A comparison of the model structure T. kodakarensis is the only euryarchaeal species that has with the previously reported structures of a family B DNA two genes encoding PCNA homologs in the genome [38]. polymerase from RB69 phage, complexed with DNA We cloned these two genes from the T. kodakarensis ge- [145,146], suggested that the second interaction site plays a nome, and the highly purified gene products, PCNA1 and crucial role in switching between the polymerase and exo- PCNA2, were characterized (Kuba et al., in review). In vivo nuclease modes, by inducing a PCNA-polymerase complex analyses revealed that the gene encoding PCNA2 could be configuration that favors synthesis over editing. This puta- knocked out, whereas no PCNA1 deletion mutants could be tive mechanism for the control of fidelity of replicative obtained, suggesting that PCNA1 is essential for DNA rep- DNA polymerases is supported by experiments in which lication, and PCNA2 may play a different role in T. koda- mutations at the second interaction site enhanced the exo- karensis. A recent report showed that both PCNA1 and nuclease activity in the presence of PCNA [146]. Further- PCNA2 form a stable ring structure and work as a proces- more, we presented the three-dimensional structure of the sivity factor for T. kodakarensis PolB in vitro. The crystal DNA polymerase-PCNA-DNA ternary complex, solved by structures of the two PCNAs revealed the different interac- electron microscopic (EM) single particle analysis [147]. tions at the subunit-subunit interfaces [153]. It will be very This structural view revealed the entire domain configura- interesting to determine how the two PCNAs function to- tion of the trimeric ring of PCNA and DNA polymerase, gether in T. kodakarensis cells. including the protein-protein or protein-DNA contacts (Fig- The eukaryotic RFC is a heteropentameric complex, con- ure 5). This architecture provides clearer insights into the sisting of five different proteins, RFC1–5, in which RFC1 is larger than the other four RFCs. Conversely, the archaeal RFC consists of two proteins, RFCS (small) and RFCL (large), in a 4:1 ratio. Thus, similar to the eukaryotic RFC, most of the archaeal RFCs form a pentameric complex. However, a different form of RFC, consisting of three sub- units, RFCS1, RFCS2, and RFCL, in a 3:1:1 ratio, was originally identified from M. acetivorans [154]. This RFC stimulates DNA synthesis by a cognate DNA polymerase in the presence of its PCNA in vitro [154]. The three subunits of RFC may represent an intermediate stage in the evolution

of the more complex RFC in Eukaryotes from the less com- Figure 5 Three-dimensional structure of the PolB-PCNA-DNA complex. plex RFC in Archaea [154,155]. The subunit organization The three-dimensional structural model of the PolB-PCNA-DNA ternary complex was obtained by EM single particle analysis, and the crystal and the spatial distribution of the subunits in the M. structure of each component was docked onto the complex structure. PolB acetivorans RFC complex were analyzed and compared interacts with PCNA primarily at two sites, a conventional PIP box and a with those of the E. coli γ-complex, which is also a pen- newly identified site (around R379). Based on this structure and the related biochemical analyses, a switching mechanism for PolB between the poly- tamer consisting of three different proteins. These two merase (synthesizing) and exonuclease (editing) modes was proposed. The clamp loaders adopt similar subunit organizations and spa- second contact between the two molecules (at R379 of PolB and E171 of tial distributions, but the functions of the individual subunits PCNA), as shown here, appears to be responsible for the configuration of are likely to be diverse [155]. Detailed evolutionary anal- the editing mode-specific complex. A different EM image with a detached second contact, indicating the synthesizing mode, was also observed (data yses of the archaeal RFC proteins, including the Mcm and not shown). PCNA proteins, were published recently [156]. 396 Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5

1.8 DNA ligase reports [172174]. The stimulatory effect of P. furiosus PCNA on the enzyme activity of the cognate DNA ligase DNA ligase is essential to connect the Okazaki fragments of was observed at a high salt concentration, at which a DNA discontinuous strand synthesis during DNA replication, and ligase alone cannot bind to a nicked DNA substrate. Fur- universally exists in all living organisms. This enzyme cat- thermore, we identified the amino acid residues that are alyzes nick-sealing reactions via three nucleotidyl transfer critical for the PCNA binding, based on mutational analyses. steps, as described in previous review articles [157,158]. In Interestingly, the binding site is located in the middle of the the first step, DNA ligase forms a covalent enzyme-AMP N-terminal DNA binding domain (DBD), and the binding intermediate, by reacting with ATP or NAD+ as a cofactor. motif, QKSFF, which we proposed as a shorter version of In the second step, DNA ligase recognizes the substrate the PIP box, is actually looped out from the protein surface. DNA, and the AMP is subsequently transferred from the We subsequently confirmed that the Gln and the first Phe in ligase to the 5′-phosphate terminus of the DNA, to form a the QKSFF motif are especially important for stable binding DNA-adenylate intermediate (AppDNA). In the final step, with PCNA [170]. Interestingly, this motif is located in the the 5′-AppDNA is attacked by the adjacent 3′-hydroxy middle of the protein chain, rather than at the N- or group of the DNA, to form a phosphodiester bond. Gener- C-terminal region, where the PIP boxes are usually found. ally, DNA ligases are grouped into two families according To confirm that this motif is conserved in the archae- to their requirement for ATP or NAD+ as a nucleotide co- al/eukaryotic DNA ligases, we showed that the physical and factor in the first reaction step. ATP-dependent DNA ligas- functional interactions between A. pernix DNA ligase and es are widely found in all three domains of life, whereas PCNA depend mainly on the phenylalanine residue, which NAD+-dependent DNA ligases exist mostly in Bacteria. is located in the region predicted by the multiple sequence NAD+-dependent enzymes are also present in halophilic alignment of the ATP-dependent DNA ligases [175]. It is archaea [159] and some eukaryotic viruses [160]. noteworthy that this shorter version of the PIP box also ex- Three genes (LIG1, LIG3 and LIG4) encoding ATP- ists in an archaeal uracil DNA glycosylase (UDG), identi- dependent DNA ligases have been identified in the human fied in our continuing work [176]. genome to date. Human DNA ligase I (LIGI), encoded by Human LIGI, complexed with DNA, was the first re- LIG1, is a replicative enzyme that joins Okazaki fragments ported crystal structure of an ATP-dependent cellular DNA during DNA replication. The first gene encoding a eukary- ligase, although the ligase was an N-terminal truncated form otic-like ATP-dependent DNA ligase was found in the [177]. The enzyme comprises the N-terminal DNA binding thermophilic archaeon Desulfolobus ambivalens [161]. domain, the middle adenylation domain, and the C-terminal Subsequent identification of DNA ligases from archaeal OB-fold domain. The crystal structure of LIGI (residues organisms revealed that these enzymes primarily use ATP 233–919), in complex with a nicked, 5'-adenylated DNA as a cofactor. However, this classification may not be so intermediate, revealed that the enzyme redirects the path of + strict; the use of NAD , as well as ATP, as a cofactor has the double-stranded DNA to expose the nick termini for the been observed in several DNA ligases, including those from strand-joining reaction. Like PCNA, the N-terminal DNA- T. kodakarensis [162], T. fumicolans, P. abyssi [163], binding domain works to encircle the DNA substrate and to Thermococcus sp. NA1 [164], T. acidophilum, Picrophilus stabilize it in a distorted structure, positioning the catalytic torridus, and Ferroplasma acidophilum [165] (Table 2). core on the nick. Our crystal structure of the full length + The dual co-factor specificity (ATP/NAD ) is interesting in DNA ligase from P. furiosus [170] revealed that the archi- terms of understanding the structures and functions of these tecture of each domain resembles those of LIGI, but the DNA ligases, although ATP is evidently preferable in all of domain arrangements differ markedly between the two en- the cases. Further dual co-factor specificity exists in the zymes. This domain rearrangement is likely derived from archaeal DNA ligases, which use both ADP and ATP, as the “domain-connecting” role of the helical extension, con- found in the enzymes from A. pernix [166] and Staphy- served at the C-termini of the archaeal and eukaryotic DNA lothermus marinus [167]. In the case of Sulfobococcus zil- ligases. The DNA substrate in the open form of LIGI is re- ligii, GTP is also the functional cofactor [168]. The DNA placed by motif VI at the C-terminus in the closed form of P. ligases from P. horikoshii [169] and P. furiosus [170] have furiosus ligase. Both the shapes and electrostatic distribu- a strict ATP preference. Sufficient data have not yet been tions are similar between motif VI and the DNA substrate, obtained to fully resolve the issue of dual co-factor specific- suggesting that motif VI in the closed state mimics the in- ity, so further biochemical and structural analyses are re- coming substrate DNA. Subsequently, another crystal quired. structure of the ATP-dependent DNA ligase from S. solfa- We determined the crystal structure of P. furiosus DNA taricus was published [178]. This crystal structure is the ligase [171] and investigated the physical and functional fully open DNA ligase structure, in which the three domains interactions between the DNA ligase and PCNA [170]. The are highly extended. In this work, the S. solfataricus lig- detailed mode of interaction between human LIGI and ase-PCNA complex was also analyzed by small-angle X-ray PCNA is somewhat unclear, because of several conflicting scattering. S. solfataricus ligase bound to the PCNA ring Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 397 retains an open, extended conformation. The closed, single PCNA ring simultaneously, because DNA ligase oc- ring-shaped conformation observed in the LIGI structure is cupies two of the three subunits of the PCNA trimer (Figure 6). probably the active form to catalyze a DNA end-joining In the case of our previous analysis of the RFC-PCNA- reaction. Therefore, it was proposed that the open-to-closed DNA complex, RFC entirely covers the PCNA ring, thus conformational change occurs for ligation, and the switch in blocking the access of other proteins (Figure 4) [140]. The the conformational change is accommodated by a malleable ternary complex appears to favor a mechanism involving interface with PCNA, which serves as an efficient platform the sequential binding and release of replication factors. In for DNA ligation [178], as illustrated in Figure 6. solution, a DNA ligase can adopt various conformations Following the publication of these crystal structures, we between the extended and closed forms, as described above. presented the three-dimensional structure of the ternary The ternary structure described by our group could be re- complex, consisting of DNA ligase-PCNA-DNA, obtained garded as an intermediate state, just prior to DNA ligation by EM single particle analysis (Figure 6) [179]. In the com- (Figure 6). plex structure, the three domains of the crescent-shaped P. furiosus ligase surround the central DNA duplex, encircled 1.9 Flap endonuclease 1 by the closed PCNA ring. The relative orientations of the ligase domains, which differ remarkably from those of the Efficient processing of Okazaki fragments is vital for DNA previous crystal structures, suggested that a large domain replication and cell proliferation. The primase-synthesized rearrangement occurs upon ternary complex formation. The RNA/DNA primers need to be removed to join the Okazaki DNA ligase contacted PCNA at two sites, the conventional fragments into an intact lagging strand DNA. Flap endonu- PIP box and a second novel contact in the middle adenyla- clease 1 (FEN1) is primarily responsible for this task. Oka- tion domain. It is also interesting that a substantial DNA tilt zaki fragment maturation is highly coordinated, and the interactions of DNA polymerase, FEN1, and DNA ligase from the PCNA ring axis was observed. Based on the struc- with PCNA allow these enzymes to act sequentially during tural model, we considered a mechanism in which the the maturation process, as described above. PCNA binding proteins are bound and released sequentially. FEN1 is a structure-specific 5′-endonuclease that specif- In fact, most of the PCNA binding proteins share the same ically recognizes a double-stranded DNA with an unan- binding sites in the interdomain connecting loop (IDCL) nealed 5′-flap [180,181]. In Eukaryota, 5′-flap DNA struc- and the C-terminal tail of the PCNA. The structural features tures are formed by the strand displacement activity of exclude the possibility that the three proteins contact the DNA polymerase δ during Okazaki fragment processing and long patch base excision repair. DNA ligase I seals the nick after the flapped DNA is cleaved by FEN1. These pro- cessing steps are facilitated by PCNA [182]. The interac- tions between eukaryotic FEN1 and PCNA have been well characterized [142,173]. The stimulatory effect of PCNA on the FEN1 activity was also examined [183]. The crystal structure of the human FEN1-PCNA complex revealed three FEN1 molecules bound to one PCNA trimer at the C-terminal PIP box (one FEN1 for each subunit), in differ- ent configurations [184]. The eukaryotic homologs of FEN1 were found in Ar- chaea [185]. Many structural analyses of archaeal FEN1 have been reported to date. The crystal structures of FEN1 from M. jannaschii [186], P. furiosus [187], P. horikoshii

[188], A. fulgidus [189], and S. solfataricus [151] have been Figure 6 Conformational changes of DNA ligase, according to the liga- determined. In addition, detailed biochemical studies were tion process. The ATP-dependent DNA ligases, consisting of three distinct domains, flexibly change their conformations to capture the DNA strand performed on P. horikoshii FEN1 [190,191]. Thus, studies and to process ligation. Three crystal structures supporting these conforma- of the archaeal FEN1 proteins have provided important in- tional changes are shown. The DNA ligases from S. solfataricus (PDB sights into the structural basis of the cleavage reaction of the code 2HIV, cyan), P. furiosus (PDB code 2CFM, green), and human LIGI flapped DNA. We cloned and purified P. furiosus FEN1, with DNA (PDB code 1X9N, dark blue and red) are shown. The predicted ligation process by DNA ligase, with the help of PCNA, is depicted at the and examined its interaction with PCNA in detail. The flap bottom. Upon binding to PCNA and DNA, the DNA ligase forms the cres- endonuclease activity of P. furiosus FEN1 was stimulated cent configuration, which embraces the DNA, as observed from the EM by PCNA. Furthermore, the stimulatory effect of PCNA on single particle analysis. The DNA ligase then grips the DNA to accomplish the sequential action of FEN1 and ligase was observed in the ligation reaction, as observed in the crystal structure of human LIGI-DNA. Accordingly, the binding sites are released, thereby enabling vitro. Based on these results, a model of the molecular the PCNA ring to interact with the PIP of the next enzyme. switching mechanism of the last steps of Okazaki-fragment 398 Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 maturation was constructed. Culture, Sports, Science, and Technology of Japan, the Japan New Energy and Industrial Technology Development Organization, and the Japan Science and Technology Agency. We apologize to the researchers whose 2 Conclusion and perspectives work was not cited because of space limitations.

Research into the molecular mechanism of DNA replication 1 Watson J D, Crick F H. Molecular structure of nucleic acids; a structure for deoxyribose nucleic acid. Nature, 1953, 171: 737–738 has been a central theme of molecular biology, and studies 2 Kelman Z, White M F. Archaeal DNA replication and repair. Curr on archaeal organisms became popular in the whole genome Opin Microbiol, 2005, 8: 669–676 sequencing age. Archaeal studies are especially interesting 3 Barry E R, Bell S D. DNA replication in the archaea. Microbiol Mol to understand the mechanisms by which cells live in ex- Biol Rev, 2006, 70: 876–887 treme environmental conditions. It is also noteworthy that 4 Wigley D B. ORC proteins: marking the start. Curr Opin Struct Biol, 2009, 19: 72–78 the proteins from the hyperthermophilic archaea are more 5 Leipe D D, Aravind L, Koonin E V. Did DNA replication evolve stable than those from mesophilic organisms, and they are twice independently? Nucleic Acids Res, 1999, 27: 3389–3401 especially advantageous for the structural and functional 6 Uemori T, Sato Y, Kato I, et al. A novel DNA polymerase in the analyses of higher-ordered complexes, such as the repli- hyperthermophilic archaeon, Pyrococcus furiosus: gene cloning, ex- pression, and characterization. Genes Cells, 1997, 2: 499–512 some. To understand each of the events involved in DNA 7 Matsunaga F, Forterre P, Ishino Y, et al. In vivo interactions of ar- metabolism, studies on the higher-order complexes, rather chaeal Cdc6/Orcl and minichromosome maintenance proteins with than single proteins, are essential. Archaeal research will the replication origin. Proc Natl Acad Sci USA, 2001, 98: 11152– continue to contribute to the development and advancement 11157 of the DNA replication research field, as summarized in a 8 Lopez P, Philippe H, Myllykallio H, et al. Identification of putative chromosomal origins of replication in Archaea. Mol Microbiol, recent review [192]. 1999, 4: 883–886 In addition to basic molecular biology research, DNA 9 Matsunaga F, Norais C, Forterre P, et al. Identification of short ‘eu- replication proteins from have been highly karyotic’ Okazaki fragments synthesized from a prokaryotic replica- useful reagents for gene manipulations, including genetic tion origin. EMBO Rep, 2003, 4:154–158 diagnosis, forensic DNA typing, detection of bacterial and 10 Robinson N P, Dionne I, Lundgren M, et al. Identification of two origins of replication in the single chromosome of the archaeon Sul- viral infections, as well as basic research. Numerous ar- folobus solfataricus. Cell, 2004, 116: 25–38 chaeal enzymes have been developed globally as commer- 11 Matsunaga F, Glatigny A, Mucchielli-Giorgi M H, et al. Ge- cial products, which are utilized daily in various processes. nomewide and biochemical analyses of DNA-binding activity of An example of the successful engineering of a DNA poly- Cdc6/Orc1 and Mcm proteins in Pyrococcus sp. Nucleic Acids Res, merase for PCR is the creation of the fusion protein of P. 2007, 35: 3214–3222 12 Liu J, Smith C L, DeRyckere D, et al. Structure and function of furiosus PolB and a nonspecific dsDNA binding protein Cdc6/Cdc18: implications for origin recognition and checkpoint from S. solfataricus, Sso7d, by genetic engineering tech- control. Mol Cell, 2000, 6: 637–648 niques [193]. To overcome the low processivity of PolB, a 13 Singleton M R, Morales R, Grainge I, et al. Conformational changes small protein with high affinity to the DNA strand was induced by nucleotide binding in Cdc6/ORC from Aeropyrum pernix. J Mol Biol, 2004, 343: 547–557 fused to the C-terminus of the enzyme. This artificial DNA 14 Grainge I, Gaudier M, Schuwirth B S, et al. Biochemical analysis of polymerase possesses high processivity, while maintaining a DNA replication origin in the archaeon Aeropyrum pernix. J Mol high fidelity for DNA synthesis. As another example, we Biol, 2006, 363: 355–369 successfully developed a novel processive PCR method, 15 Dueber E L, Corn J E, Bell S D, et al. Replication origin recognition using archaeal PolB with the help of a mutant PCNA and deformation by a heterodimeric archaeal Orc1 complex. Science, 2007, 317: 1210–1213 [194,195]. Improved DNA sequencing technologies, re- 16 Gaudier M, Schuwirth B S, Westcott, S L, et al. Structural basis of ferred to as “next-generation sequencing” have been devel- DNA replication origin recognition by an ORC protein. Science, oped [196,197] and single-molecule detection, using 2007, 317: 1213–1216 dye-labeled modified nucleotides and longer read lengths, 17 Majernik A I, Chong I P. A conserved mechanism for replication known as “third-generation DNA sequencing” [198] is also origin recognition and binding in archaea. Biochem J, 2008, 409: 511–518 available. All of these technologies include DNA polymer- 18 Matsunaga F, Takemura K, Akita M, et al. Localized melting of du- ases or DNA ligases from various sources including archae- plex DNA by Cdc6/Orc1 at the DNA replication origin in the hy- al organisms, indicating that DNA replication enzymes are perthermophilic archaeon Pyrococcus furiosus. Extremophiles, indispensable for the development of DNA manipulation 2009, 14: 21–31 19 Akita M, Adachi A, Takemura K, et al. Cdc6/Orc1 from Pyrococcus technology. Further efforts will provide valuable enzymes furiosus may act as the origin recognition protein and Mcm helicase with specialized activities for promoting genetic engineer- recruiter. Gens Cells, 2010, 15: 537–552 ing technologies, and thermostable enzymes with their as- 20 Lundgren M, Andersson A, Chen L, et al. Three replication origins sociated factors have great potential. in Sulfolobus species: synchronous initiation of chromosome repli- cation and asynchronous termination. Proc Natl Acad Sci USA, 2004, 101: 7046–7051 Our work cited in this review was supported in part by the Human Frontier 21 Robinson N P, Blood K A, McCallum S A, et al. Sister chromatid Science Program, several research grants from Ministry of Education, junctions in the hyperthormophilic archaeaon Sulfolobus solfataricus. Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 399

EMBO J, 2007, 26: 816–824 Thermococcus kodakarensis. Genes Cells, 2011, 16: 1176–1189 22 Robinson N P, Bell S D. Extrachromosomal element capture and the 43 Li Z, Santangelo T J, Čuboňová L, et al. Affinity purification of an evolution of multiple replication origins in archaeal chromosomes. archaeal DNA replication protein network. mBio, 2010, 1: Proc Natl Acad Sci USA, 2007, 104: 5806–5811 e00221-10 23 Norais C, Hawkins M, Hartman A L, et al. Genetic and physical 44 Fujikane R, Ishino S, Ishino Y, et al. Genetic analysis of DNA repair mapping of DNA replication origins in Haloferax volcanii. PLoS in the hyperthermophilic archaeon, Thermococcus kodakarensis. Genet, 2007, 3:e77 Genes Genet Syst, 2010, 85: 243–257 24 Capes M D, Coker J A, Gessler R, et al. The information transfer 45 Xia Q, Hendrickson E L, Zhang Y, et al. Quantitative proteomics of system of halophilic archaea. Plasmid, 2011, 65: 77–101 the archaeon Methanococcus maripaludis validated by microarray 25 Masai H, You Z, Arai K. Control of DNA replication: regulation and analysis and real time PCR. Mol Cell Proteomics, 2006, 5: 868–881 activation of eukaryotic replicative helicase, MCM. IUBMB Life, 46 Walters A D, Chong J P. An archaeal order with multiple mini- 2005, 57: 323–335 chromosome maintenance genes. Microbiology, 2010, 156: 1405– 26 Ilves I, Petojevic T, Pesavento J J, et al. Activation of the MCM2-7 1414 helicase by association with Cdc45 and GINS proteins. Mol Cell, 47 Takayama Y, Kamimura Y, Okawa M, et al. GINS, a novel multi- 2010, 37: 247–258 protein complex required for chromosomal DNA replication in bud- 27 Chong J P, Hayashi M K, Simon M N, et al. A double-hexamer ar- ding yeast. Genes Dev, 2003, 17: 1153–1165 chaeal minichromosome maintenance protein is an ATP-dependent 48 Makarova K S, Wolf Y I, Mekhedov S L, et al. Ancestral paralogs DNA helicase. Proc Natl Acad Sci USA, 2000, 97: 1530–1535 and pseudoparalogs and their role in the emergence of the eukaryotic 28 Grainge I, Scaife S, Wigley D B. Biochemical analysis of compo- cell. Nucleic Acids Res, 2005, 33: 4626–4638 nents of the pre-replication complex of Archaeoglobus fulgidus. Nu- 49 Marinsek N, Barry E R, Makarova K S, et al. GINS, a central nexus cleic Acids Res, 2003, 31: 4888–4898 in the archaeal DNA replication fork. EMBO Rep, 2006, 7: 539–545 29 Kelman Z, Lee J K, Hurwitz J. The single minichromosome mainte- 50 Pacek M, Tutter A V, Kubota Y, et al. Localization of MCM2-7, nance protein of Methanobacterium thermoautotrophicum ∆H con- Cdc45, and GINS to the site of DNA unwinding during eukaryotic tains DNA helicase activity. Proc Natl Acad Sci USA, 1999, 96: DNA replication. Mol Cell, 2006, 21: 581–587 14783–14788 51 Ogino H, Ishino S, Mayanagi K, et al. The GINScomplex from the 30 Shechter D F, Ying C Y, Gautier J. The intrinsic DNA helicase ac- thermophilic archaeon, Thermoplasma acidophilummay function as tivity of Methanobacterium thermoautotrophicum ∆H minichromo- a homotetramer in DNA replication. Extremophiles, 2011, 15: some maintenance protein. J Biol Chem, 2000, 275: 15049–15059 529–539 31 Yoshimochi T, Fujikane R, Kawanami M, et al. The GINS complex 52 Oyama T, Ishino S, Fujino S, et al. Architectures of archaeal GINS from Pyrococcus furiosus stimulates the MCM helicase activity. J complexes, essential DNA replication initiation factors. BMC Biol, Biol Chem, 2008, 283: 1601–1609 2011, 9: 28 32 Sakakibara N, Kelman L M, Kelman Z. Unwinding the structure and 53 Kamada K, Kubota Y, Arata T, et al. Structure of the human GINS function of the archaeal MCM helicase. Mol Microbiol, 2009, 72: complex and its assembly and functional interface in replication ini- 286–296 tiation. Nat Struct Mol Biol, 2007, 14: 388–396 33 Shin J H, Grabowski B, Kasiviswanathan R, et al. Regulation of 54 Choi J M, Lim H S, Kim J J, et al. Crystal structure of the human minichromosome maintenance helicase activity by Cdc6. J Biol GINS complex. Genes Dev, 2007, 21: 1316–1321 Chem, 2003, 278: 38059–38067 55 Chang Y P, Wang G, Bermudez V, et al. Crystal structure of the 34 Kasiviswanathan R, Shin J H, Kelman Z. Interactions between the GINS complex and functional insight into its role in DNA replica- archaeal Cdc6 and MCM proteins modulate their biochemical prop- tion. Proc Natl Acad Sci USA, 2007, 104: 12685–12690 erties. Nucleic Acids Res, 2005, 33: 4940–4950 56 Sanchez-Pulido L, Ponting C P. Cdc45: the missing RecJ ortholog in 35 De Felice M, Esposito L, Pucci B, et al. Biochemical characteriza- eukaryotes? Bioinformatics, 2011, 27: 1885–1888 tion of a CDC6-like protein from the crenarchaeon Sulfolobus solfa- 57 Li Z, Pan M, Santangelo T J, et al. A novel DNA nuclease is stimu- taricus. J Biol Chem, 2003, 278: 46424–46431 lated by association with the GINS complex. Nucleic Acids Res, 36 Haugland G T, Shin J H, Birkeland N K, et al. Stimulation of MCM 2011, 39: 6114–6123 helicase activity by a Cdc6 protein in the archaeon Thermoplasma 58 Krastanova I, Sannino V, Amenitsch H, et al. Structural and func- acidophilum. Nucleic Acids Res, 2006, 34: 6337–6344 tional insights into the DNA replication factor Cdc45 reveal an evo- 37 Krupovic M, Gribaldo S, Bamford D H, et al. The evolutionary his- lutionary relationship to the DHH family of phosphoesterases. J Biol tory of archaeal MCM helicases: a case study of vertical evolution Chem, 2012, 287: 4121–4128 combined with hitchhiking of mobile genetic elements. Mol Biol 59 Kirk B W, Kuchta R D. Arg304 of human DNA primase is a key Evol, 2010, 27: 2716–2732 contributor to catalysis and NTP binding: primase and the family X 38 Fukui T, Atomi H, Kanai T, et al. Complete genome sequence of the polymerases share significant sequence homology. Biochemistry, hyperthermophilic archaeon Thermococcus kodakarensis KOD1 and 1999, 38: 7727–7736 comparison with Pyrococcus genomes. Genome Res, 2005, 15: 60 Desogus G, Onesti S, Brick P, et al. Identification and characteriza- 352–363 tion of a DNA primase from the hyperthermophilic archaeon Meth- 39 Sato T, Fukui T, Atomi H, et al. Targeted gene disruption by ho- anococcus jannaschii. Nucleic Acids Res, 1999, 27: 4444–4450 mologous recombination in the hyperthermophilic archaeon Ther- 61 Bocquier A, Liu L, Cann I, et al. Archaeal primase: bridging the gap mococcus kodakaraensis KOD1. J Bacteriol, 2003, 185: 210–220 between RNA and DNA polymerase. Curr Biol, 2001, 11: 452-456 40 Sato T, Fukui T, Atomi H, et al. Improved and versatile transfor- 62 Liu L, Komori K, Ishino S, et al. The archaeal DNA primase: bio- mation system allowing multiple genetic manipulations of the hy- chemical characterization of the p41-p46 complex from Pyrococcus perthermophilic archaeon Thermococcus kodakaraensis. Appl En- furiosus. J Biol Chem, 2001, 276: 45484–45490 viron Microbiol, 2005, 71: 3889–3899 63 Lao-Sirieix S H, Bell S D. The heterodimeric primase of the hyper- 41 Pan M, Santangelo T J, Li Z, et al. Thermococcus kodakarensis en- thermophilic archaeon Sulfolobus solfataricus possesses DNA and codes three MCM homologs but only one is essential. Nucleic Acids RNA primase, polymerase and 3'-terminal nucleotidyl transferase Res, 2011, 39: 9671–9680 activities. J Mol Biol, 2004, 344: 1251–1263 42 Ishino S, Fujino S, Tomita H, et al. Biochemical and genetical anal- 64 Lao-Sirieix S H, Nookala R K, Roversi P, et al. Structure of the het- yses of the three mcm genes from the hyperthermophilic archaeon, erodimeric core primase. Nat Struct Mol Biol, 2005, 12: 1137–1144 400 Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5

65 De Falco M, Fusco A, DeFelice M, et al. The DNA primase is acti- 85 Kenny M K, Lee S H, Hurwitz J. Multiple functions of human sin- vated by substrates containing a thymine-rich bubble and has a gle-stranded-DNA binding protein in simian virus 40 DNA replica- 3'-terminal nucleotidyl-transferase activity. Nucleic Acids Res, 2004, tion: single-strand stabilization and stimulation of DNA polymerases 32: 5223–5230 alpha and delta. Proc Natl Acad Sci USA, 1989, 86: 9757–9761 66 Ito N, Nureki O, Shirouzu M, et al. Crystal structure of the Pyro- 86 Chedin F, Seitz E M, Kowalczykowski S C. Novel homologs of rep- coccus horikoshii DNA primase-UTP complex: implications for the lication protein A in archaea: implications for the evolution of mechanism of primer synthesis. Genes Cells, 2003, 8: 913–923 ssDNA-binding proteins. Trends Biol Sci, 1998, 23: 273–277 67 Matsui E, Nishio M, Yokoyama H, et al. Distinct domain functions 87 Kelly T J, Simancek P, Brush G S. Identification and characteriza- regulating de novo DNA synthesis of thermostable DNA primase tion of a single-stranded DNA-binding protein from the archaeon from hyperthermophile Pyrococcus horikoshii. Biochemistry. 2003, Methanococcus jannaschii. Proc Natl Acad Sci USA, 1998, 95: 42: 14968–14976 14634–14639 68 Ito N, Matsui I, Matsui E. Molecular basis for the subunit assembly 88 Kelman Z, Pietrokovski S, Hurwitz J. Isolation and characterization of the primase from an archaeon Pyrococcus horikoshii. FEBS J, of a split B-type DNA polymerase from the archaeon Methanobac- 2007, 274: 1340–1351 terium thermoautotrophicum ∆ H. J Biol Chem, 1999, 274: 69 Le Breton M, Henneke G, Norais C, et al. The heterodimeric pri- 28751–28761 mase from the euryarchaeon Pyrococcus abyssi: a multifunctional 89 Komori K, Ishino Y. Replication protein A in Pyrococcus furiosus is enzyme for initiation and repair? J Mol Biol, 2007, 374: 1172–1185 involved in homologous DNA recombination. J Biol Chem, 2001, 70 Hu J, Guo L, Wu K, et al. Template-dependent polymerization 276: 25654–25660 across discontinuous templates by the heterodimeric primase from 90 Kerr I D, Wadsworth R I, Cubeddu L, et al. Insights into ssDNA the hyperthermophilic archaeon Sulfolobus solfataricus. The Nucleic recognition by the OB fold from a structural and thermodynamic Acids Research advance online publication 20 December 2011; doi: study of Sulfolobus SSB protein. EMBO J, 2003, 22: 2561–2570 10.1093/nar/gkr1256 91 Robbins J B, Murphy M C, White B A, et al. Functional analysis of 71 Evguenieva-Hackenberg E, Walter P, Hochleitner E, et al. An exo- multiple single-stranded DNA-binding proteins from Methano- some-like complex in Sulfolobus solfataricus. EMBO Rep, 2003, 4: sarcina acetivorans and their effects on DNA synthesis by DNA 889–893 polymerase BI. J Biol Chem, 2004, 279: 6315–6326 72 Walter P, Klein F, Lorentzen E, et al. Characterization of native and 92 Lin Y, Lin L J, Sriratana P, et al. Engineering of functional replica- reconstituted exosome complexes from the hyperthermophilic ar- tion protein a homologs based on insights into the evolution of oli- chaeon Sulfolobus solfataricus. Mol Microbiol, 2006, 62: 1076– gonucleotide/oligosaccharide-binding folds. J Bacteriol, 2008, 190: 1089 5766–5780 73 Zuo Z, Rodgers C J, Mikheikin A L,et al. Characterization of a 93 Braithwaite D K, Ito J. Compilation, alignment, and phylogenetic functional DnaG-type primase in archaea: implications for a du- relationships of DNA polymerases. Nucleic Acids Res, 1993, 21: al-primase system. J Mol Biol, 2010, 397: 664–676 787–802 74 Wu K, Lai X, Guo X, et al. Interplay between primase and replica- 94 Cann I, Ishino Y. Archaeal DNA replication: Identifying the pieces tion factor C in the hyperthermophilic archaeon Sulfolobus solfata- to solve a puzzle. Genetics, 1999, 152: 1249–1267 ricus. Mol Microbiol, 2007, 63: 826–837 95 Ishino Y, Cann I. The euryarchaeotes, a subdomain of archaea, sur- 75 Murzin A G. OB(oligonucleotide/oligosaccharide binding)-fold: vive on a single DNA polymerase: fact or farce? Genes Genet Syst, common structural and functional solution for non-homologous se- 1998, 73: 323–336 quences. EMBO J, 1993, 12: 861–867 96 Lipps G, Röther S, Hart C, et al. A novel type of replicative enzyme 76 Braun K A, Lao Y, He Z, et al. Wold MS Role of protein-protein harbouring ATPase, primase and DNA polymerase activity. EMBO interactions in the function of replication protein A (RPA): RPA J, 2003, 22: 2516–2525 modulates the activity of DNA polymerase alpha by multiple mech- 97 Ohmori H, Friedberg E C, Fuchs R P, et al. The Y-family of DNA anisms. Biochemistry, 1997, 36: 8443–8454 polymerases. Mol Cell, 2001, 8: 7–8 77 Lin Y L, Chen C, Keshav K F, et al. Dissection of functional do- 98 Guo C, Kosarek-Stancel J N, Tang T S, et al. Y-family DNA poly- mains of the human DNA replication protein complex replication merases in mammalian cells. Cell Mol Life Sci, 2009, 66: 2363– protein A. J Biol Chem, 1996, 271: 17190–17198 2381 78 Bochkarev A, Bochkareva E, Frappier L, et al. The crystal structure 99 Uemori T, Ishino Y, Doi H, et al. The hyperthermophilic archaeon of the complex of replication protein A subunits RPA32 and RPA14 Pyrodictium occultum has two alpha-like DNA polymerases. J Bac- reveals a mechanism for single-stranded DNA binding. EMBO J, teriol, 1995, 177: 2164–2177 1999, 18: 4498–4504 100 Cann I, Ishino S, Nomura N, et al. Two family B DNA polymerases 79 Bochkareva E, Frappier L, Edwards A M, et al. The RPA32 subunit in Aeropyrum pernix, an obligate aerovic hyperthermophilic crenar- of human replication protein A contains a single-stranded chaeote. J Bacteriol, 1999, 181: 5984–5992 DNA-binding domain. J Biol Chem, 1998, 273: 3932–3936 101 Cann I, Komori K, Toh H, et al. A heterodimeric DNA polymerase: 80 Philipova D, Mullen J R, Maniar H S, et al. A hierarchy of SSB evidence that members of the euryarchaeota possess a novel DNA protomers in replication protein A. Genes Dev, 1996, 10: 2222–2233 polymerase. Proc Natl Acad Sci USA, 1998, 95: 14250–14255 81 Gomes X V, Wold M S. Structural analysis of human replication 102 Ishino S, Ishino Y. Conprehensive search for DNA polymerase in protein A. Mapping functional domains of the 70-kD subunit. J Biol the hyperthermophilic archaeon, Pyrococcus furiosus. Nucleos Nu- Chem, 1995, 270: 4534–4543 cleot Nucleic Acids, 2006, 25: 681–691 82 Mer G, Bochkarev A, Gupta R, et al. Structural basis for the recog- 103 Uemori T, Ishino Y, Toh H, et al. Organization and nucleotide se- nition of DNA repair proteins UNG2, XPA, and RAD52 by replica- quence of the DNA polymerase gene from the archaeon Pyrococcus tion factor RPA. Cell, 2000, 103: 449–456 furiosus. Nucleic Acids Res, 1992, 21: 259–265 83 Dornreiter I, Erdile L F, Gilbert I U, et al. Interaction of DNA pol- 104 Perler F B, Kumar S, Kong H. Thermostable DNA polymerases. ymerase alpha-primase with cellular replication protein A and SV40 Adv Protein Chem, 1996, 48: 377–435 T antigen. EMBO J, 1992, 11: 769–776 105 Imamura K, Fukunaga K, Kawarabayasi Y, et al. Specific interac- 84 Tsurimoto T, Stillman B. Multiple replication factors augment DNA tions of three PCNAs with replication-related proteins in Aeropyrum synthesis by the two eukaryotic DNA polymerases, alpha and delta. pernix. Mol Microbiol, 2007, 64: 308–318 EMBO J, 1989, 8: 3883–3889 106 Ishino Y, Ishino S. Novel DNA polymerases from Euryarchaeota. Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 401

Meth Enzymol, 2001, 334: 249–260 lication fork. Cell, 2007, 129, 665–679 107 Gueguen Y, Rolland J L, Lecompte O, et al. Characterization of two 129 Pan M, Kelman L, Kelman Z. The archaeal PCNA proteins. Bio- DNA polymerases from the hyperthermophilic euryarchaeon Pyro- chem Soc Trans, 2011, 39: 20–24 coccus abyssi. Eur J Biochem, 2001, 268: 5961–5969 130 Cann I, Ishino S, Hayashi I, et al. Functional interactions of a hom- 108 Shen Y, Musti K, Hiramoto M, et al. Invariant Asp-1122 and olog of proliferating cell nuclear antigen with DNA polymerases in Asp-1124 are essential residues for polymerization catalysis of fam- Archaea. J Bacteriol, 1999, 181: 6591–6599 ily D DNA polymerase from Pyrococcus horikoshii. J Biol Chem, 131 Ishino Y, Tsurimoto T, Ishino S, et al. Functional interactions of an 2001, 276: 27376–27383 archaeal sliding clamp with mammalian clamp loader and DNA 109 Shen Y, Tang X, Matsui I. Subunit interaction and regulation of ac- polymerase δ. Genes Cells, 2001, 6: 699–706 tivity through terminal domains of the family D DNA polymerase 132 Matsumiya S, Ishino S, Ishino Y, et al. Physical interaction between from Pyrococcus horikoshii. J Biol Chem, 2003, 278: 21247–21257 proliferating cell nuclear antigen and replication factor C from Py- 110 Tang X F, Shen Y, Matsui E, et al. Domain topology of the DNA rococcus furiosus. Genes Cells, 2002, 7: 911–922 polymerase D complex from a hyperthermophilic archaeon Pyro- 133 Matsumiya S, Ishino S, Ishino Y, et al. Intermolecular ion pairs coccus horikoshii. Biochemistry, 2004, 43: 11818–11827 maintain toroidal structure of Pyrococcus furiosus PCNA. Prot Sci, 111 Jokela M, Eskelinen A, Pospiech H, et al. Characterization of the 3' 2003, 12: 823–831 exonuclease subunit DP1 of Methanococcus jannaschii replicative 134 Tori K, Kimizu M, Ishino S, et al. Both DNA polymerase BI and D DNA polymerase D. Nucleic Acids Res, 2004, 32: 2430–2440 from the hyperthermophilic archaeon, Pyrococcus furiosus bind to 112 Henneke G, Flament D, Hübscher U, et al. The hyperthermophilic PCNA at the C-terminal PIP box motifs. J Bacteriol, 2007, 189: euryarchaeota Pyrococcus abyssi likely requires the two DNA pol- 5652–5657 ymerases D and B for DNA replication. J Mol Biol, 2005, 350: 135 Cann I, Ishino S, Yuasa M, et al. Biochemical analysis of the repli- 53–64 cation factor C from the hyperthermophilic archaeon Pyrococcus fu- 113 Castrec B, Laurent S, Henneke G, et al. The glycine-rich motif of riosus. J Bacteriol, 2001, 183: 2614–2623 Pyrococcus abyssi DNA polymerase D is critical for protein stability. 136 Mayanagi K, Miyata T, Oyama T, et al. Three-dimensional electron J Mol Biol, 2010, 396: 840–848 microscopy of clamp loader small subunit from Pyrococcus furiosus. 114 Berquist B R, DasSarma P, DasSarma S. Essential and non-essential J Struct Biol, 2001, 134: 35–45 DNA replication genes in the model halophilic Archaeon, Halobac- 137 Ishino S, Oyama T, Yuasa M, et al. Mutational analysis of Pyro- terium sp. NRC-1. BMC Genet, 2007, 8:31 coccus furiosus replication factor C based on the three-dimensional 115 Brochier-Armanet C, Boussau B, Gribaldo S, et al. Mesophilic structure. Extremophiles, 2003, 7: 169–175 Crenarchaeota: proposal for a third archaeal phylum, the Thaumar- 138 Oyama T, Ishino Y, Cann I, et al. Atomic structure of the clamp chaeota. Nat Rev Microbiol, 2008, 6: 245–252 loader small subunit from Pyrococcus furiosus. Mol Cell, 2001, 8: 116 Nunoura T, Takaki Y, Kakuta J, et al. Insights into the evolution of 455–463 Archaea and eukaryotic protein modifier systems revealed by the 139 Indiani C, O’Donnell M. The replication clamp-loading machine at genome of a novel archaeal group. Nucleic Acids Res, 2011, 39: work in the three domains of life. Nat Rev Mol Cell Biol, 2006, 7: 3204–3223 751–761 117 Elkins J G, Podar M, Graham D E, et al. A korarchaeal genome re- 140 Miyata T, Suzuki H, Oyama T, et al. Open clamp structure in the veals insights into the evolution of the Archaea. Proc Natl Acad Sci clamp-loading complex visualized by electron microscopic image USA, 2008, 105: 8102–8107 analysis. Proc Natl Acad Sci USA, 2005, 102: 13795–13800 118 Schleper C, Swanson R V, Mathur E J, et al. Characterization of a 141 Kelch B A, Makino D L, O’Donnell M, et al. The replication DNA polymerase from the uncultivated psychrophilic archaeon clamp-loading machine at work in the three domains of life. Science, Cenarchaeum symbiosum. J Bacteriol, 1997, 179: 7803–7811 2011, 334: 1675–1680 119 Tahirov T H, Makarova K S, Rogozin I B, et al. Evolution of DNA 142 Warbrick E M. The puzzle of PCNA’s many partners. BioEssays, polymerases: an inactivated polymerase-exonuclease module in Pol 2000, 22: 997–1006 epsilon and a chimeric origin of eukaryotic polymerases from two 143 Vivona J B, Kelman Z. The diverse spectrum of sliding clamp inter- classes of archaeal ancestors. Biol Direct, 2009, 4: 11 acting proteins. FEBS Lett, 2003, 546: 167–167 120 Connolly B A. Recognition of deaminated bases by archaeal fami- 144 Nishida H, Mayanagi K, Kiyonari S, et al. Structural determinant for ly-B DNA polymerases. Biochem Soc Trans, 2009, 37: 65–68 switching between the polymerase and exonuclease modes in the 121 Wardle J, Burgers P M, Cann I K, et al. Uracil recognition by repli- PCNA-replicative DNA polymerase complex. Proc Natl Acad Sci cative DNA polymerases is limited to the archaea, not occurring USA, 2009, 106: 20693–20698 with bacteria and eukarya. Nucleic Acids Res, 2008, 36: 705–711 145 Shamoo Y, Steitz T A. Building a replisome from interacting pieces: 122 Fogg M G, Pearl L H, Conolly B A. Structural basis for uracil sliding clamp complexed to a peptide from DNA polymerase and a recognition by archaeal family B DNA polymerases. Nat Struct Biol, polymerase editing complex. Cell, 1999, 99: 155–166 2002, 9: 922–927 146 Franklin M C, Wang J, Steitz T A. Structure of the replicating com- 123 Zillig W, Kletzin A, Schleper C, et al. Screening for Sulfolobales, plex of a pol alpha family DNA polymerase. Cell, 2001, 105: their plasmids and their viruses in Icelandic solfataras. Syst Appl 657–667 Microbiol, 1993, 16: 609–628 147 Mayanagi K, Kiyonari S, Nishida H, et al. Architecture of the DNA 124 Lipps G, Röther S, Hart C, et al. A novel type of replicative enzyme polymerase B-proliferating cell nuclear antigen (PCNA)-DNA ter- harbouring ATPase, primase and DNA polymerase activity. EMBO nary complex. Proc Natl Acad Sci USA, 2011, 108: 1845–1849 J, 2003, 22: 2516–2525 148 Daimon K, Kawarabayasi Y, Kikuchi H, et al. Three proliferating 125 Lipps G. Molecular biology of the pRN1 plasmid from Sulfolobus cell nuclear antigen-like proteins found in the hyperthermophilic ar- islandicus. Biochem Soc Trans, 2009, 37: 42–45 chaeon Aeropyrum pernix: interactions with the two DNA polymer- 126 Lipps G. The replication protein of the Sulfolobus islandicus plas- ases. J Bacteriol, 2002, 184: 687–694 mid pRN1. Biochem Soc Trans, 2004, 32: 240–244 149 Dionne I, Nookala R K, Jackson S P, et al. A heterotrimeric PCNA 127 Soler N, Marguet E, Cortez D, et al. Two novel families of plasmids in the hyperthermophilic archaeon Sulfolobus solfataricus. Mol Cell, from hyperthermophilic archaea encoding new families of replica- 2003,11: 275–282 tion proteins. Nucleic Acids Res, 2010, 38: 5088–5104 150 Imamura K, Fukunaga K, Kawarabayasi Y, et al. Specific interac- 128 Moldovan G L, Pfander B, Jentsch S. PCNA, the maestro of the rep- tions of three proliferating cell nuclear antigens with replication- 402 Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5

related proteins in Aeropyrum pernix. Mol Microbiol, 2007, 64: 172 Levin D S, Bai W, Yao N, et al. An interaction between DNA ligase 308–318 I and proliferating cell nuclear antigen: implications for Okazaki 151 Williams G J, Johnson K, Rudolf J, et al. Structure of the heterotri- fragment synthesis and joining. Proc Natl Acad Sci USA, 1997, 94: meric PCNA from Sulfolobus solfataricus. Acta Crystallogr Sect F 12863–12868 Struct Biol Cryst Commun, 2006, 62: 944–948 173 Jónsson Z O, Hindges R, Hübscher U. Regulation of DNA replica- 152 Doré A S, Kilkenny M L, Jones S A, et al. Structure of an archaeal tion and repair proteins through interaction with the front side of PCNA1-PCNA2-FEN1 complex: elucidating PCNA subunit and proliferating cell nuclear antigen. EMBO J, 1998, 17: 2412–2425 client enzyme specificity. Nucleic Acids Res, 2006, 34: 4515–4526 174 Tom S, Henricksen L A, Park M S, et al. DNA ligase I and prolifer- 153 Ladner J E, Pan M, Hurwitz J, et al. Crystal structures of two active ating cell nuclear antigen form a functional complex. J Biol Chem, proliferating cell nuclear antigens (PCNAs) encoded by Thermo- 2001, 276: 24817–24825 coccus kodakaraensis. Proc Natl Acad Sci USA, 2011, 108: 2711– 175 Kiyonari S, Kamigochi T, Ishino Y. A single amino acid substitution 2716 in the DNA-binding domain of Aeropyrum pernix DNA ligase im- 154 Chen Y H, Kocherginskaya S A, Lin Y, et al. Biochemical and mu- pairs its interaction with proliferating cell nuclear antigen. Ex- tational analyses of a unique clamp loader complex in the archaeon tremophiles, 2007, 11: 675–684 Methanosarcina acetivorans. J Biol Chem, 2005, 280: 41852–41863 176 Kiyonari S, Uchimura M, Shirai T, et al. Physical and functional in- 155 Chen Y H, Lin Y, Yoshinaga A, et al. Molecular analyses of a teractions between uracil-DNA glycosylase and proliferating cell three-subunit euryarchaeal clamp loader complex from Methano- nuclear antigen from the euryarchaeon Ptrococcus furiosus. J Biol sarcina acetivorans. J Bacteriol, 2009, 191: 6539–6549 Chem, 2008, 283: 24185–24193 156 Chia N, Cann I, Olsen G J. Evolution of DNA replication protein 177 Pascal J M, O’Brien P J, Tomkinson A E, et al. Human DNA ligase complexes in eukaryotes and Archaea. PLoS ONE, 2010, 5: e10866 I completely encircles and partially unwinds nicked DNA. Nature, 157 Wilkinson A, Day J, Bowater R. Bacterial DNA ligases. Mol Micro- 2004, 432: 473–478 biol, 2001, 40: 1241–1248 178 Pascal J M, Tsodikov O V, Hura G L, et al. A flexible interface be- 158 Tomkinson A E, Vijayakumar S, Pascal J M, et al. DNA ligases: tween DNA ligase and PCNA supports conformational switching structure, reaction mechanism, and function. Chem Rev, 2006, 106: and efficient ligation of DNA. Mol Cell, 2006, 24: 279–291 687–699 179 Mayanagi K, Kiyonari S, Saito M, et al. Mechanism of replication 159 Poidevin L, MacNeill S A. Biochemical characterisation of LigN, an machinery assembly as revealed by the DNA ligase-PCNA-DNA NAD+-dependent DNA ligase from the halophilic euryarchaeon complex architecture. Proc Natl Acad Sci USA, 2009, 106: 4657– Haloferax volcanii that displays maximal in vitro activity at high salt 4652 concentrations. BMC Mol Biol, 2006, 7: 44 180 Liu Y, Kao H I, Bambara R A. Flap endonuclease 1: a central com- 160 Benarroch D, Shuman S. Characterization of mimivirus NAD+- ponent of DNA metabolism. Annu Rev Biochem, 2004, 73: 589–615 dependent DNA ligase. Virology, 2006, 353: 133–143 181 Zheng L, Jia J, Finger L D, et al. Functional regulation of FEN1 nu- 161 Kletzin A. Molecular characterisation of a DNA ligase gene of the clease and its link to cancer. Nucleic Acids Res, 2011, 39: 781–794 extremely thermophilic archaeon Desulfurolobus ambivalens shows 182 Rossi M L, Bambara R A. Reconstituted Okazaki fragment pro- close phylogenetic relationship to eukaryotic ligases. Nucleic Acids cessing indicates two pathways of primer removal. J Biol Chem, Res, 1992, 20: 5389–5396 2006, 281: 26051–26061 162 Nakatani M, Ezaki S, Atomi H, et al. A DNA ligase from a hyper- 183 Tom S, Henricksen L A, Bambara R A. Mechanism whereby prolif- thermophilic archaeon with unique cofactor specificity. J Bacteriol, erating cell nuclear antigen stimulates flap endonuclease 1. J Biol 2000, 182: 6424–6433 Chem, 2000, 275: 10498–10505 163 Rolland J L, Gueguen Y, Persillon C, et al. Characterization of a 184 Sakurai S, Kitano K, Yamaguchi H, et al. Structural basis for re- thermophilic DNA ligase from the archaeon Thermococcus fumi- cruitment of human flap endonuclease 1 to PCNA. EMBO J, 2005, colans. FEMS Microbiol Lett, 2004, 236: 267–273 24: 683–693 164 Kim Y J, Lee H S, Bae S S., et al. Cloning, expression, and charac- 185 Shen B, Qiu J, Hosfield D, et al. Flap endonuclease homologs in ar- terization of a DNA ligase from a hyperthermophilic archaeon chaebacteria exist as independent proteins. Trends Biochem Sci, Thermococcus sp. Biotechnol Lett, 2006, 28: 401–407 1998, 23: 171–173 165 Ferrer M, Golyshina O V, Beloqui A, et al. A purple acidophilic 186 Hwang K Y, Baek K, Kim H Y, et al. The crystal structure of flap di-ferric DNA ligase from Ferroplasma. Proc Natl Acad Sci USA, endonuclease-1 from Methanococcus jannaschii. Nat Struct Biol, 2008, 105: 8878–8883 1998, 5: 707–713 166 Jeon S J, Ishikawa K. A novel ADP-dependent DNA ligase from 187 Hosfield D J, Mol C D, Shen B, et al. Structure of the DNA repair Aeropyrum pernix K1. FEBS Lett, 2003, 550: 69–73 and replication endonuclease and exonuclease FEN-1: coupling 167 Seo M S, Kim Y J, Choi J J, et al. Cloning and expression of a DNA DNA and PCNA binding to FEN-1 activity. Cell, 1998, 95: 135–146 ligase from the hyperthermophilic archaeon mari- 188 Matsui E, Musti K V, Abe J, et al. Molecular structure and novel nus and properties of the enzyme. J Biotechnol, 2007, 128: 519–530 DNA binding sites located in loops of flap endonuclease-1 from 168 Sun Y, Seo M S, Kim J H, et al. Novel DNA ligase with broad nu- Pyrococcus horikoshii. J Biol Chem, 2002, 277: 37840–37847 cleotide cofactor specificity from the hyperthermophilic crenar- 189 Chapados B R, Hosfield D J, Han S, et al. Structural basis for FEN-1 chaeon Sulfophobococcus zilligii: influence of ancestral DNA ligase substrate specificity and PCNA-mediated activation in DNA replica- on cofactor utilization. Environ Microbiol, 2008, 10: 3212–3224 tion and repair. Cell, 2004, 116: 39–50 169 Keppetipola N, Shuman S. Characterization of a thermophilic 190 Matsui E, Kawasaki S, Ishida H, et al. Thermostable flap endonu- ATP-dependent DNA ligase from the euryarchaeon Pyrococcus clease from the archaeon, Pyrococcus horikoshii, cleaves the repli- horikoshii. J Bacteriol, 2005, 187: 6902–6908 cation fork-like structure endo/exonucleolytically. J Biol Chem, 170 Kiyonari S, Takayama K, Nishida H, et al. Identification of a novel 1999, 274: 18297–182309 binding motif in Pyrococcus furiosus DNA ligase for the functional 191 Matsui E, Abe J, Yokoyama H, et al. Aromatic residues located interaction with proliferating cell nuclear antigen. J Biol Chem, close to the active center are essential for the catalytic reaction of 2006, 281: 28023–28032 flap endonuclease-1 from hyperthermophilic archaeon Pyrococcus 171 Nishida H, Kiyonari S, Ishino Y, et al. The closed structure of an horikoshii. J Biol Chem, 2004, 279:16687–16696 archaeal DNA ligase from Pyrococcus furiosus. J Mol Biol, 2006, 192 Beattie T R, Bell S D. Molecular machines in archaeal DNA replica- 360: 956–967 tion. Curr Opin Chem Biol, 2011, 15: 614–619 Ishino Y, et al. Sci China Life Sci May (2012) Vol.55 No.5 403

193 Wang Y, Prosen D E, Mei L, et al. A novel strategy to engineer 199 Jackson B R, Noble C, Lavesa-Curto M, et al. Characterization of an DNA polymerases for enhanced processivity and improved perfor- ATP-dependent DNA ligase from the acidophilic archaeon “Ferro- mance in vitro. Nucleic Acids Res, 2004, 32: 1197–1207 plasma acidarmanus” Fer1. Extremophiles, 2007, 11: 315–327 194 Kawamura A, Ishino Y, Ishino S. Biophysical analysis of PCNA 200 Sriskanda V, Kelman Z, Hurwitz J, et al. Characterization of an from Pyrococcus furiosus. J Jap Soc Extremophiles, 2012, in press ATP-dependent DNA ligase from the thermophilic archaeon Meth- 195 Ishino S, Kawamura A, Ishino Y. Application of PCNA to proces- anobacterium thermoautotrophicum. Nucleic Acids Res, 2000, 28: sive PCR by reducing the stability of its ring structure. J Jap Soc 2221–2228 Extremophiles, 2012, in press 201 Lai X, Shao H, Hao F, et al. Biochemical characterization of an 196 Shendure J, Ji H. Next-generation DNA sequencing. Nat Biotechnol, ATP-dependent DNA ligase from the hyperthermophilic crenar- 2008, 26: 1135–1145 chaeon Sulfolobus shibatae. Extremophiles, 2002, 6: 469–477 197 Ansorge W J. Next-generation DNA sequencing techniques. N Bio- 202 Bezsudnova E Y, Kovalchuk M V, Mardanov A V, et al. Overex- technol, 2009, 25: 195–203 pression, purification and crystallization of a thermostable DNA 198 Metzker M L. Sequencing technologies - the next generation. Nat ligase from the archaeon Thermococcus sp. 1519. Acta Crystallogr Rev Genet, 2010, 11: 31–46 Sect F Struct Biol Cryst Commun, 2009, 65: 368–371

Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.