718
Views
0
CrossRef citations to date
0
Altmetric
EDITORIALS: CELL CYCLE FEATURES

FEN1-mediated α-segment error editing during Okazaki fragment maturation

, &
Page 1767 | Received 21 Apr 2015, Accepted 27 Apr 2015, Published online: 17 Jun 2015

DNA-replication-repair machinery determines the non-random occurrence of variations across the genome. During eukaryotic nuclear-genome replication, most parts of the leading and lagging strand are synthesized by high fidelity DNA polymerase ε and δ respectively. However, a third enzyme polymerase (Pol) α-primase initiates each synthesis event. During Okazaki fragment, down-stream of the RNA primer and synthesis, DNA fragment synthesized by proofreading-deficient Pol-α, which we termed as α-segment, provide major source of mutations and undergo DNA repair process to remove the incorporated errors.Citation1 The α-segment contributes ∼1.5% of total genome. A series of the very recent papers made available by Nature press indicates that despite Okazaki fragment processing, DNA synthesized by Pol α is retained in vivo.Citation2-5 DNA-binding proteins including histones and transcription factors that rapidly re-associate, post-replication, to act as partial barriers to Pol-δ-mediated displacement of the α-segment, resulting increased mutation rates in the region. What is the cellular mechanism that minimizes the mutation frequency in the alpha-segment? Limited evidence is available. Pol α variant strains have mild mutator phenotypes, resulting from DNA replication errors generated by Pol α, and their mutation rates are synergistically increased by loss of MSH2-dependent mismatch repair (MMR). Thus, MSH2-dependent MMR in yeast plays a major role in correcting mismatches in the α-segment.Citation1

Additional study indicated that some mismatches generated by yeast Pol α are excised by exonuclease 1 (EXO1), a 5′ exonuclease involved in MMR.Citation6 Interestingly, the loss of MMR resulting from deleting EXO1 is mild compared to the loss of MMR from deleting MSH2, implying the existence of a MSH2-dependent but EXO1-independent mechanism that removes mismatches from the α-segment. In our paper currently in press with EMBO Journal,Citation7 we investigated the molecular mechanism mediated by FEN1 and MSH2 functional complex to edit the errors in the α-segment. Our unique substrate designing strategy segregates the RNA primer removal (RPR) and α-segment error editing (AEE) activities of FEN1 and characterize efficiency of functional enzymatic component of nuclear extract (NEs). Interestingly, besides its well-characterized RNA primer removl function, FEN1 was able to go on in an exonuclease fashion in presence of the mis-paired nucleotides in the α-segment and remove them within a limited distance (12 nts) from the nick MSH2 interacts with FEN1 and facilitates its nuclease activity to remove mismatches near the 5′ ends of DNA substrates. Mouse cells and mice encoding FEN1 AEE defective mutations display a strong mutator phenotype, enhanced cellular transformation, and increased cancer susceptibility.Citation7

The proposed AEE mechanism is significant because it effectively removes replication errors while concomitantly maturing Okazaki fragments, which is a rate-limiting step in DNA replication.Citation7 However, if a mutation occurs in AEE components that deregulates the process and leads to constitutive editing and futile cycles, it could affect DNA ligation and result in spontaneous DNA strand breaks and consequently, various forms of chromosome aberrations. Such a mutation would be different from the ones due to defects in RPR process, of which the major form is the duplication mutation. Both the mutations that result in DNA strand breaks and aneuploidy and the duplication mutation, are hallmarks of human cancer. The current study identifies a novel role of FEN1 in a specialized mismatch repair pathway and a new cancer etiological mechanism.

References

  • McElhinny NSA, et al. Proc Natl Acad Sci U S A 2010; 107:21070-5; PMID: 21041657; http://dx.doi.org/10.1073/pnas.1013048107
  • Reijns MA, Kemp H, Ding J, de Proce SM, Jackson AP, Taylor MS. Lagging-strand replication shapes the mutational landscape of the genome. Nature 2015; 518:502-6; PMID: 25624100; http://dx.doi.org/10.1038/nature14183
  • Koh KD, Balachander S, Hesselberth JR, Storici F. Ribose-seq: global mapping of ribonucleotides embedded in genomic DNA. Nature methods 2015; 12:251-7; PMID: 25622106; http://dx.doi.org/10.1038/nmeth.3259
  • Daigaku Y, Keszthelyi A, Muller CA, Miyabe I, Brooks T, Retkute R, Hubank M, Nieduszynski CA, Carr AM. A global profile of replicative polymerase usage. Nature structural & molecular biology. Nat Commun 2015; 22:192–8; PMID: 25664722; http://dx.doi.org/10.1038/nsmb.2962
  • Clausen AR, et al. Nat Struct Mol Biol 2015; 22:185-91; PMID:25622295; http://dx.doi.org/10.1038/nsmb.2957
  • Liberti SE, et al. DNA Repair (Amst) 2013; 12:92-6; PMID:23245696; http://dx.doi.org/10.1016/j.dnarep.2012.11.001
  • Liu S, et al. The EMBO J 2015; PMID: 25921062; http://dx.doi.org/10.15252/embj.201489865

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.