首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
The mechanism by which cells recognize and complete replicated regions at their precise doubling point must be remarkably efficient, occurring thousands of times per cell division along the chromosomes of humans. However, this process remains poorly understood. Here we show that, in Escherichia coli, the completion of replication involves an enzymatic system that effectively counts pairs and limits cellular replication to its doubling point by allowing converging replication forks to transiently continue through the doubling point before the excess, over-replicated regions are incised, resected, and joined. Completion requires RecBCD and involves several proteins associated with repairing double-strand breaks including, ExoI, SbcDC, and RecG. However, unlike double-strand break repair, completion occurs independently of homologous recombination and RecA. In some bacterial viruses, the completion mechanism is specifically targeted for inactivation to allow over-replication to occur during lytic replication. The results suggest that a primary cause of genomic instabilities in many double-strand-break-repair mutants arises from an impaired ability to complete replication, independent from DNA damage.During chromosomal replication, cells tightly regulate the processes of initiation, elongation, and completion to ensure that each daughter cell inherits an identical copy of the genetic information. Although the mechanisms regulating initiation and elongation have been well characterized (reviewed in refs. 1, 2), the process of how cells recognize replicated regions and complete replication at the precise doubling point remains a fundamental question yet to be addressed. Whether this event occurs once per generation as in Escherichia coli or thousands of times per generation as in human cells, the failure to efficiently carry out this function would be expected to result in a loss of genomic stability. Considering the large number of proteins that cells devote to ensuring the fidelity of replication initiation and elongation, it seems highly probable that the final critical step in this process will be also be tightly regulated and controlled enzymatically.In some aspects, one could argue that the efficiency of completion is likely to be more critical to the faithful duplication of the genome than that of initiation. When replication origins fail to initiate efficiently, elongation of replication forks from neighboring origins is often able to compensate (3, 4), and both prokaryotic and eukaryotic cells are able to tolerate variations in their origin number without severe phenotypic consequences (57). However, a failure to accurately limit or join any event where forks converge would be expected to result in duplications, deletions, rearrangements, or a loss of viability depending upon how the DNA ends are resolved at segregation.A number of studies suggest that an ability to sense when all sequences in the genome have doubled is critical to genomic replication. In vitro, converging replisomes continue through their meeting point as one replisome displaces the other, resulting in over-replication, or a third copy, of the region where the forks meet (8). Complicating the process of genomic doubling even further, several studies have suggested that illegitimate initiations of replication frequently occur at single-strand nicks, gaps, D-loops, and R-loops throughout the genomes of both prokaryotes and eukaryotes (914). Similar to when replication forks continue through a previously replicated template, each of these events would generate a third copy of the chromosomal region where the event occurs. Thus, over-replication may be inherent and promiscuous during the duplication of genomes. If true, then to ensure that each sequence of the genome replicates once, and only once, per generation, cells must encode an enzymatic system that is essentially able to count in pairs and efficiently degrade odd or over-replicated regions until the two nascent end pairs of replication events can be joined.The model organism E. coli is particularly well-suited to dissect how this fundamental process occurs. In E. coli, the completion of replication occurs at a defined region on the genome, opposite to the bidirectional origin of replication (15). Most completion events can be further localized to one of six termination (ter) sequences within the 400-kb terminus region due to the action of Tus, which binds to ter and inhibits replication fork progression in an orientation-dependent manner, in effect stalling the replication fork at this site until the second arrives (16, 17). Although Tus confines converging replication forks to a specific region, it does not appear to be directly involved in the completion reaction because tus mutants have no phenotype and complete replication normally (18). Furthermore, plasmids and bacteriophage lacking ter sequences are maintained stably (19).Many mutants impaired for either replication initiation or elongation were initially isolated based on their growth defects or an impaired ability to maintain plasmids (2022). We reasoned that mutants impaired for the ability to complete replication might be expected to exhibit similar phenotypes and initially focused our attention on the properties of recBC and recD mutants. RecB-C-D forms a helicase–nuclease complex that is required for homologous repair of double-strand breaks in E. coli (23, 24). The enzyme uses specific DNA sequences, termed “Chi sites,” to initiate recombination between pairs of molecules. Loss of RecB or C inactivates the enzyme complex, whereas loss of RecD inactivates the nuclease and Chi recognition, but retains helicase activity (23, 24). Here, we show that inactivation of RecBCD leads to a failure to recognize and join replicating molecules at their doubling point. Although the completion process requires RecBCD, it is distinct from double-strand break repair and does not involve a double-strand break intermediate, homologous recombination, or RecA.  相似文献   

2.
Repair of a chromosomal double-strand break (DSB) by gene conversion depends on the ability of the broken ends to encounter a donor sequence. To understand how chromosomal location of a target sequence affects DSB repair, we took advantage of genome-wide Hi-C analysis of yeast chromosomes to create a series of strains in which an induced site-specific DSB in budding yeast is repaired by a 2-kb donor sequence inserted at different locations. The efficiency of repair, measured by cell viability or competition between each donor and a reference site, showed a strong correlation (r = 0.85 and 0.79) with the contact frequencies of each donor with the DSB repair site. Repair efficiency depends on the distance between donor and recipient rather than any intrinsic limitation of a particular donor site. These results further demonstrate that the search for homology is the rate-limiting step in DSB repair and suggest that cells often fail to repair a DSB because they cannot locate a donor before other, apparently lethal, processes arise. The repair efficiency of a donor locus can be improved by four factors: slower 5′ to 3′ resection of the DSB ends, increased abundance of replication protein factor A (RPA), longer shared homology, or presence of a recombination enhancer element adjacent to a donor.Homologous recombination is the predominant mechanism to repair chromosome breaks and preserve genome integrity. In eukaryotes, the broken double-strand break (DSB) ends undergo extensive 5′ to 3′ resection, promoting the binding of the Rad51 recombinase to form a nucleoprotein filament that can search the genome for a homologous sequence with which it can effect repair. Donor template sequences can be located on a sister chromatid, a homologous chromosome or an ectopic location. When ectopic sequences are used, repair results in nonallelic replacements of sequences. In budding yeast Saccharomyces cerevisiae it is possible to monitor the sequence of DSB repair events in real time by Southern blots, PCR or chromatin immunoprecipitation (1).Haploid yeast chromosomes are arranged in a Rabl orientation, with the 16 centromeres all clustered at the spindle-pole body (SPB) whereas the telomeres are associated in loose clusters at the nuclear envelope (2). These observations have been extended by the use of chromosome conformation capture approaches (3, 4) to map the relative positions of loci along each chromosome based on their frequencies of crosslinking (contact frequencies) with many other sites in the genome. Previous studies have shown that telomere-associated sequences preferentially recombine with other telomere-associated loci whereas centromere-linked sites selectively recombine with other centromere-linked loci (57). However, such preferences, presumably caused by the constraints of tethering, may not reflect the general behavior of most sequences undergoing homologous recombination. It is not known how the position of the sequences, lying between the tethered ends, influences their ability to repair a DSB.Taking advantage of the Hi-C database, we established a series of yeast strains in which a DSB, induced at a defined location, could be repaired by a short, homologous donor sequence whose location in each strain was chosen for its apparent contact frequency with the recipient. In this way we were able to learn how the relative positions of sequences influences their recombinational potential and whether, as seems to be the case for translocations arising in mammalian cells (8, 9), chromosomal proximity influences the efficiency or rate of repair. We find that there is a strong correlation between the 3D position of a donor sequence within the yeast nucleus and its efficiency in repairing a DSB and that a given locus has very different repair potential depending on the location of the chromosome break.  相似文献   

3.
Recombinational DNA repair by the RecF pathway of Escherichia coli requires the coordinated activities of RecA, RecFOR, RecQ, RecJ, and single-strand DNA binding (SSB) proteins. These proteins facilitate formation of homologously paired joint molecules between linear double-stranded (dsDNA) and supercoiled DNA. Repair starts with resection of the broken dsDNA by RecQ, a 3′→5′ helicase, RecJ, a 5′→3′ exonuclease, and SSB protein. The ends of a dsDNA break can be blunt-ended, or they may possess either 5′- or 3′-single-stranded DNA (ssDNA) overhangs of undefined length. Here we show that RecJ nuclease alone can initiate nucleolytic resection of DNA with 5′-ssDNA overhangs, and that RecQ helicase can initiate resection of DNA with blunt-ends or 3′-ssDNA overhangs by DNA unwinding. We establish that in addition to its well-known ssDNA exonuclease activity, RecJ can display dsDNA exonuclease activity, degrading 100–200 nucleotides of the strand terminating with a 5′-ssDNA overhang. The dsDNA product, with a 3′-ssDNA overhang, is an optimal substrate for RecQ, which unwinds this intermediate to reveal the complementary DNA strand with a 5′-end that is degraded iteratively by RecJ. On the other hand, RecJ cannot resect duplex DNA that is either blunt-ended or terminated with 3′-ssDNA; however, such DNA is unwound by RecQ to create ssDNA for RecJ exonuclease. RecJ requires interaction with SSB for exonucleolytic degradation of ssDNA but not dsDNA. Thus, complementary action by RecJ and RecQ permits initiation of recombinational repair from all dsDNA ends: 5′-overhangs, blunt, or 3′-overhangs. Such helicase–nuclease coordination is a common mechanism underlying resection in all organisms.Homologous recombination is a relatively error-free mechanism to repair double-stranded DNA (dsDNA) breaks (DSBs) and single-stranded DNA (ssDNA) gaps, which are produced by UV light, γ-irradiation, and chemical mutagens (1). In wild-type Escherichia coli, the labor of recombinational repair is divided between the RecBCD and RecF pathways of recombination, which are responsible for the repair of DSBs and ssDNA gaps, respectively (25). However, the proteins of the RecF pathway are capable of DSB repair, as well as ssDNA gap repair: in recBC mutant cells containing the suppressor mutations, sbcB and sbcC (suppressors of recBC), the proteins of the RecF pathways provide the needed recombinational DNA repair functions (2, 6).The RecF pathway in E. coli involves the functions of RecA, RecF, RecG, RecJ, RecN, RecO, RecQ, RecR, RuvA, RuvB, RuvC, and single-strand DNA binding (SSB) proteins (1, 7). The RecF pathway of recombination is evolutionarily conserved across Bacteria, with most of components present in all bacteria (8). In addition, orthologs of RecF pathway proteins are found in Eukarya. RecA promotes DNA strand invasion and exchange (911), as does eukaryotic Rad51 (12, 13). RecO can both anneal SSB–ssDNA complexes (14, 15) and, in conjunction with RecR (and RecF), mediate loading of RecA onto SSB–ssDNA complexes (1618). Saccharomyces cerevisiae Rad52 is a functional homolog of RecO in that it also displays both DNA-annealing and Rad51-loading activities (1922). The RecFOR complex promotes the loading of RecA onto SSB-coated gapped DNA at ssDNA–dsDNA junctions (17, 18) and, when mutated, is suppressed by hyperactive alleles of recA (23), a property that is shared with the yeast Rad55/57 proteins (24). Furthermore, human BRCA2 protein and a fungal analog, Brh2, are partial functional analogs of the RecFOR proteins (2527).RecQ helicase plays several roles in both early and late steps of recombination (28, 29), as do the RecQ-family helicases in Eukarya [e.g., Sgs1 and Bloom Syndrome helicase (BLM)] (3032). In addition, eukaryotic Exonuclease 1 (Exo1) and Dna2 helicase/nuclease function somewhat analogously, although not identically, to RecJ nuclease (3336). The in vitro reconstitution of DSB repair in E. coli, yeast, and human have shown that resection involves specific pairs of a helicase and nuclease for DNA end resection: RecQ/RecJ, Sgs1/Dna2, BLM/DNA2, and BLM/EXO1 (28, 3739).A comparison of DSB repair by the RecBCD and RecF pathways shows that repair starts with the processing a DSB into resected dsDNA with a 3′-ssDNA overhang (7). RecJ has a 5′ to 3′ exonuclease activity on ssDNA and the action of RecJ is facilitated by RecQ, which has a 3′ to 5′ helicase activity (40, 41). The resulting processed DNA has a 3′-ssDNA overhang. The RecFOR complex binds to the 5′-end at the junction between ssDNA and dsDNA, and loads RecA protein onto the adjacent ssDNA (17, 18). Finally, the RecA nucleoprotein filament promotes pairing with homologous dsDNA (9). These steps have been reconstituted in vitro in a coordinated reaction using RecAFORQJ and SSB proteins (28).Despite progress, most studies have used DNA substrates with simple blunt-ends. However, in vivo, there are many potential structures at the end of a DSB. When the DSB is created by a replication fork encountering nicked DNA, the break can be blunt-ended (5). However, related mechanisms can produce dsDNA with either 5′- or 3′-ssDNA overhangs. Similarly, the actual intermediates of DNA processing may result in dsDNA with either 5′- or 3′-ssDNA ends. Clearly, a DNA repair pathway must be capable of dealing with such a variety of DNA end structures. In this study, we investigated the processing of DSBs by RecJ and RecQ, both individually and together. We found that a DNA with a 5′-ssDNA overhang end was degraded by RecJ nuclease and converted into an intermediate with a 3′-ssDNA overhang. Although this intermediate was no longer a substrate for RecJ, RecQ could bind to this intermediate and initiate unwinding, thereby supplying 5′-tailed ssDNA for further resection by RecJ. In addition, we established that RecQ allows RecJ to initiate nucleolytic resection on otherwise poor substrates (e.g., blunt-end DNA or DNA with 3′-ssDNA overhangs). Thus, RecQ and RecJ cooperate biochemically to create DNA intermediates for one another that enable resection of all types of broken DNA molecules.  相似文献   

4.
5.
Hereditary cases of breast and ovarian cancer are often attributed to germ-line mutations of the BRCA1 tumor suppressor gene. Although BRCA1 is involved in diverse cellular processes, its role in the maintenance of genomic integrity may be a key component of its tumor suppression activity. The protein encoded by BRCA1 interacts in vivo with the related BARD1 protein to form a heterodimeric complex that acts as a ubiquitin E3 ligase. Because the enzymatic activity of the BRCA1/BARD1 heterodimer is conserved over a broad phylogenetic range, it is thought to be critical for the central functions of BRCA1. To test this hypothesis, we have generated isogenic clones of embryonic stem cells that do or do not express an enzymatically proficient Brca1 polypeptide. Surprisingly, cells lacking the ubiquitin ligase activity of BRCA1 are viable and do not accumulate spontaneous cytogenetic rearrangements. Gene targeting efficiencies are modestly reduced in these cells, and chromosomal rearrangements arise at elevated rates in response to genotoxic stress. Nonetheless, cells lacking Brca1 enzymatic activity are not hypersensitive to the DNA cross-linking agent mitomycin C. They also form Rad51 focus in response to ionizing radiation and repair chromosome breaks by homologous recombination at wild-type levels. These results indicate that key aspects of BRCA1 function in genome maintenance, including its role in homology-directed repair of double-strand DNA breaks, do not depend on the E3 ligase activity of BRCA1.  相似文献   

6.
The classical nonhomologous DNA end-joining (C-NHEJ) double-strand break (DSB) repair pathway in mammalian cells maintains genome stability and is required for V(D)J recombination and lymphocyte development. Mutations in the XLF C-NHEJ factor or ataxia telangiectasia-mutated (ATM) DSB response protein cause radiosensitivity and immunodeficiency in humans. Although potential roles for XLF in C-NHEJ are unknown, ATM activates a general DSB response by phosphorylating substrates, including histone H2AX and 53BP1, which are assembled into chromatin complexes around DSBs. In mice, C-NHEJ, V(D)J recombination, and lymphocyte development are, at most, modestly impaired in the absence of XLF or ATM, but are severely impaired in the absence of both. Redundant functions of XLF and ATM depend on ATM kinase activity; correspondingly, combined XLF and H2AX deficiency severely impairs V(D)J recombination, even though H2AX deficiency alone has little impact on this process. These and other findings suggest that XLF may provide functions that overlap more broadly with assembled DSB response factors on chromatin. As one test of this notion, we generated mice and cells with a combined deficiency for XLF and 53BP1. In this context, 53BP1 deficiency, although leading to genome instability, has only modest effects on V(D)J recombination or lymphocyte development. Strikingly, we find that combined XLF/53BP1 deficiency in mice severely impairs C-NHEJ, V(D)J recombination, and lymphocyte development while also leading to general genomic instability and growth defects. We conclude that XLF is functionally redundant with multiple members of the ATM-dependent DNA damage response in facilitating C-NHEJ and discuss implications of our findings for potential functions of these factors.  相似文献   

7.
8.
The insertion of DNA into a genome can result in the duplication and dispersal of functional sequences through the genome. In addition, a deeper understanding of insertion mechanisms will inform methods of genetic engineering and plant transformation. Exploiting structural variations in numerous rice accessions, we have inferred and analyzed intermediate length (10–1,000 bp) insertions in plants. Insertions in this size class were found to be approximately equal in frequency to deletions, and compound insertion–deletions comprised only 0.1% of all events. Our findings indicate that, as observed in humans, tandem or partially tandem duplications are the dominant form of insertion (48%), although short duplications from ectopic donors account for a sizable fraction of insertions in rice (38%). Many nontandem duplications contain insertions from nearby DNA (within 200 bp) and can contain multiple donor sources—some distant—in single events. Although replication slippage is a plausible explanation for tandem duplications, the end homology required in such a model is most often absent and rarely is >5 bp. However, end homology is commonly longer than expected by chance. Such findings lead us to favor a model of patch-mediated double-strand-break creation followed by nonhomologous end-joining. Additionally, a striking bias toward 31-bp partially tandem duplications suggests that errors in nucleotide excision repair may be resolved via a similar, but distinct, pathway. In summary, the analysis of recent insertions in rice suggests multiple underappreciated causes of structural variation in eukaryotes.Genomic DNA insertion causes genome expansion and, potentially, the rearrangement and diffusion of protein domains and regulatory elements throughout the genome (1, 2). Additionally, genetic engineers generally aim to integrate specific DNA into the nuclear genome, so the natural mechanisms by which this integration occurs may serve as a starting point to elaborate and improve genome modification (3, 4). Common causes of gene-sized insertions are unequal recombination (5), transposable element replication (1), and ectopic recombination stimulated by double-strand breaks (DSBs) in the genome (2, 6). Shorter events are less well characterized, but it appears that these can be created by similar processes (7). Still, high-throughput sequencing of DSB repair events in humans (8) and plants (9) suggests that insertions related to induced breaks are very rare and very short.Although the processes described above can produce duplications at distant genetic loci, the most common form of non-microsatellite-associated insertions in humans is tandem duplications (10). Once created, tandem duplications can be dramatically expanded by unequal recombination or replication slippage. Such duplications may be deleterious, or they may be promoted by selection for a novel or expanded function (11, 12).Although tandem repeats are ubiquitous in eukaryotic genomes, the mechanisms for their origin are still in question. Early analysis of human indel mutations indicated that replication slippage was the most effective model to explain the origin of assorted repeats (13). In other studies, longer, de novo tandem duplications were also hypothesized to be caused by slippage because, out of 85 insertions producing such duplications, 50 were associated with flanking repeats >2 bp (14). Replication slippage would presumably require a preexisting short repeat because priming must occur between the end of the loop that will become the duplication and the position to where replication slips. Authors of more recent work investigating insertions across the human genome suggest alternatives to replication slippage on the grounds that homology is often either nonexistent or very short, whereas the length of homology and the length of insertion are not correlated (10). These researchers favor a model based on DSBs being repaired by nonhomologous end-joining (NHEJ). However, conventional models of DSB repair are strained to predict tandem duplications >10 bp, much less >100 bp. Such models require extensive single-stranded, complementary ends to be preserved during the break. Moreover, DSBs produced by Tal-effector nucleases in humans do not yield insertions that form tandem repeats, despite the fact that the breaks generate a 5′ overhang (15). Thus, this common class of mutations currently lacks a firm molecular explanation.Similar to tandem duplications, short duplications are commonly found within 100 bp of one another, but with unique intervening DNA (16). By comparing human polymorphisms with chimp sequence, Thomas et al. (16) inferred that the repeats were recent insertions. As discussed by the authors and herein, a mechanism for such duplications is even less forthcoming than for tandem duplications.In this study, we used extensive population-scale rice resequencing data to confirm that tandem duplications are also abundant natural polymorphisms in the plant kingdom. Additionally, we found that many insertions in rice, although not perfectly tandem, are from a ∼50-bp window around the insertion site. We rarely found the end homology in tandem repeats that is expected for replication slippage, although we did note a bias toward short microhomology between insertion ends and insertion site. These data led us to elaborate on the DSB model of tandem duplication, proposing that long patch base excision repair (BER) on complementary strands commonly leads to such patterns (17). Additionally, we characterized common forms of nontandem, but local, duplication.  相似文献   

9.
Poly(ADP-ribose) polymerase (PARP) inhibitors are strikingly toxic to cells with defects in homologous recombination (HR). The mechanistic basis for these findings is incompletely understood. Here, we show that PARP inhibitor treatment induces phosphorylation of DNA-dependent protein kinase substrates and stimulates error-prone nonhomologous end joining (NHEJ) selectively in HR-deficient cells. Notably, inhibiting DNA-dependent protein kinase activity reverses the genomic instability previously reported in these cells after PARP inhibition. Moreover, disabling NHEJ by using genetic or pharmacologic approaches rescues the lethality of PARP inhibition or down-regulation in cell lines lacking BRCA2, BRCA1, or ATM. Collectively, our results not only implicate PARP1 catalytic activity in the regulation of NHEJ in HR-deficient cells, but also indicate that deregulated NHEJ plays a major role in generating the genomic instability and cytotoxicity in HR-deficient cells treated with PARP inhibitors.  相似文献   

10.
11.
Nonhomologous end-joining (NHEJ) is the major DNA double-strand break (DSB) repair pathway in mammals and resolves the DSBs generated during both V(D)J recombination in developing lymphocytes and class switch recombination (CSR) in antigen-stimulated B cells. In contrast to the absolute requirement for NHEJ to resolve DSBs associated with V(D)J recombination, DSBs associated with CSR can be resolved in NHEJ-deficient cells (albeit at a reduced level) by a poorly defined alternative end-joining (A-EJ) pathway. Deletion of DNA ligase IV (Lig4), a core component of the NHEJ pathway, reduces CSR efficiency in a mouse B-cell line capable of robust cytokine-stimulated CSR in cell culture. Here, we report that CSR levels are not further reduced by deletion of either of the two remaining DNA ligases (Lig1 and nuclear Lig3) in Lig4−/− cells. We conclude that in the absence of Lig4, Lig1, and Lig3 function in a redundant manner in resolving switch region DSBs during CSR.DNA double-strand breaks (DSBs) are one of the most severe forms of DNA damage that can result from pathological conditions such as replication stress, exposure to ionizing radiation (IR), free radicals, or other DNA-damaging drugs or because of failed single-strand break repair (SSBR) (1, 2). In developing lymphocytes, programmed DSBs are essential intermediates for antigen receptor gene rearrangements, including V(D)J recombination and Ig heavy chain class switch recombination (CSR) (1, 2). Homologous recombination (HR) and nonhomologous end-joining (NHEJ) are the two major pathways for DSB repair. Whereas HR is restricted to the S/G2 phase of the cell cycle, NHEJ is active throughout the cell cycle and is generally considered the major pathway for DSB repair in mammals (1, 2).The NHEJ pathway has been extensively studied. The core components include the Ku70/Ku86 heterodimer, DNA-dependent protein kinase, X-ray cross complementation factor 4 (XRCC4), and DNA ligase IV (Lig4) (1, 2). Additional NHEJ factors include the Artemis nuclease, XRCC4-like factor (XLF) (or Cernunnos), Paralog of XRCC4 and XLF, and Polymerases µ and λ. Missing any of these factors results in various degrees of DSB repair deficits that are highly context-dependent. In general, cells lacking core components of NHEJ are hypersensitive to IR and abolished for V(D)J recombination but are only partially defective for CSR and competent for circulation of transfected linearized plasmids, suggesting that there exists an “alternative” way to join at least some types of DSBs. This alternative end-joining (A-EJ) pathway has recently become a focal area of research because of its implications in oncogenic chromosomal translocations (3), which are rare in NHEJ-proficient cells but much more frequent when NHEJ is compromised. Little is known about A-EJ other than it is kinetically slow and uses an increased level of microhomology (nucleotide overlaps that can be assigned to either of the two DNA ends) during joining (2, 4). A number of DNA repair factors, many of which are involved in SSBR, have been implicated in A-EJ (5), but the overall composition of A-EJ remains elusive. It is still unclear whether A-EJ is a distinct pathway, consists of multiple subpathways, or is merely an aberrant form of NHEJ with missing components substituted by compatible but less efficient factors. It is also unclear whether A-EJ contributes to DSB repair in NHEJ-proficient cells at all or is only active when NHEJ is compromised.Much of our understanding of mechanistic details of DSB repair has derived from studies of V(D)J recombination and CSR; both involving DSB intermediates (1). V(D)J recombination is initiated by the recombination-activating genes (RAGs) that bind and cleave at specific DNA sequences flanking the V, D, and J segments to assemble an exon encoding the variable (antigen binding) domain of the B- and T-cell receptors. CSR is initiated by activation-induced cytidine deaminase (AID) in antigen-stimulated B cells that changes the IgH constant (C) region to a different isotype. AID catalyzes DNA cytosine deamination (converting cytosines to uracils) at switch regions preceding each C region (6, 7). Processing of AID-generated uracils, through a mechanism still not fully characterized, leads to DSB formation. Although both processes use NHEJ to join DSBs, in cells missing any of the core components of NHEJ, CSR is only partially defective, whereas V(D)J recombination is completely abolished. It has been reported that the RAG complex holds the four broken ends in a postcleavage complex and directs VDJ-associated DSBs into the NHEJ pathway (8, 9). In contrast, significant levels of CSR can occur in the absence of any core NHEJ factors (10, 1114), suggesting that switch region breaks are more accessible to alternative DSB repair pathways.Regardless of how broken DNA ends are processed, at least one DNA ligase is required to ligate the two ends. Vertebrates have three ATP-dependent DNA ligases (Lig1, Lig3, and Lig4) (15). Lig1 and Lig4 are conserved in all eukaryotes, whereas Lig3 is only present in vertebrates (15). Lig4 is a core component of the NHEJ pathway and functions exclusively in NHEJ. Cells deficient for Lig4, or its cofactor XRCC4, display the most severe phenotypes of NHEJ deficiency. In the absence of Lig4, A-EJ must rely on Lig1 or Lig3 (or both). It is generally accepted that the major role of Lig1 is to join Okazaki fragments during DNA replication, and this function is mediated by an interaction with the proliferating cell nuclear antigen (PCNA) (16). Lig3 is produced in somatic cells in two forms (mitochondrial and nuclear) via alternative translation initiation (17). It was recently shown that mitochondrial, but not nuclear, Lig3 is essential for cell viability (18, 19). Nuclear Lig3 stably interacts with the X-ray complementation factor 1 (XRCC1), a scaffold protein that is essential for base excision repair. For this reason, Lig3 is regarded as the primary DNA repair ligase for SSBR, although Lig1 has also been implicated in DNA repair (15).We have previously reported the disruption Lig4 and Lig1 in a mouse B-cell line (CH12F3) capable of robust cytokine-induced CSR (10). Lig4−/− CH12F3 cells undergo kinetically slow but significant levels of CSR [∼50% of wild-type (WT) level at day 3] (10). Switch junctions isolated from Lig4−/− CH12F3 cells show increased microhomology and no direct joins (10). In the present study, we focused on determining which of two remaining ligases (Lig1 and Lig3) is responsible for A-EJ during CSR in Lig4−/− cells. To that end, we disrupted Lig1 and Lig3 (nuclear) individually in Lig4−/− CH12F3 cells. We found that Lig1 and Lig3 have redundant functions in DNA repair in response to a variety of DNA-damaging agents and during repair by A-EJ during CSR.  相似文献   

12.
Single-stranded DNA (ssDNA) covered with the heterotrimeric Replication Protein A (RPA) complex is a central intermediate of DNA replication and repair. How RPA is regulated to ensure the fidelity of DNA replication and repair remains poorly understood. Yeast Rtt105 is an RPA-interacting protein required for RPA nuclear import and efficient ssDNA binding. Here, we describe an important role of Rtt105 in high-fidelity DNA replication and recombination and demonstrate that these functions of Rtt105 primarily depend on its regulation of RPA. The deletion of RTT105 causes elevated spontaneous DNA mutations with large duplications or deletions mediated by microhomologies. Rtt105 is recruited to DNA double-stranded break (DSB) ends where it promotes RPA assembly and homologous recombination repair by gene conversion or break-induced replication. In contrast, Rtt105 attenuates DSB repair by the mutagenic single-strand annealing or alternative end joining pathway. Thus, Rtt105-mediated regulation of RPA promotes high-fidelity replication and recombination while suppressing repair by deleterious pathways. Finally, we show that the human RPA-interacting protein hRIP-α, a putative functional homolog of Rtt105, also stimulates RPA assembly on ssDNA, suggesting the conservation of an Rtt105-mediated mechanism.

Faithful DNA replication and repair are essential for the maintenance of genetic material (1). Even minor defects in replication or repair can cause high loads of mutations, genome instability, cancer, and other diseases (1). Deficiency in different DNA repair or replication proteins can lead to distinct mutation patterns (24). For example, deficiency in mismatch repair results in increased microsatellite instability, while deficiency in homologous recombination repair is often associated with tandem duplications or deletions (37). Sequence analysis of various cancer types has identified many distinct genome rearrangement and mutation signatures (8). However, the genetic basis for some of these signatures remains poorly understood, thus requiring further investigation in experimental models (8).In eukaryotic cells, Replication Protein A (RPA), the major single-stranded DNA (ssDNA) binding protein complex, is essential for DNA replication, repair, and recombination (913). It is also crucial for the suppression of mutations and genome instability (1417). RPA acts as a key scaffold to recruit and coordinate proteins involved in different DNA metabolic processes (14, 15, 17). As the first responder of ssDNA, RPA participates in both replication initiation and elongation (10, 12, 13). During replication or under replication stresses, the exposed ssDNA must be protected and stabilized by RPA to prevent formation of secondary structures (14, 16). RPA is also essential for DNA double-stranded break (DSB) repair by the homologous recombination (HR) pathway (1821). During HR, the 5′-terminated strands of DSBs are initially processed by the resection machinery, generating 3′-tailed ssDNA (22). The 3′-ssDNA becomes bound by the RPA complex to activate the DNA damage checkpoint (23). RPA is subsequently replaced by the Rad51 recombinase to form a Rad51 nucleoprotein filament (19, 24). This recombinase filament catalyzes invasion of the 3′-strands at the homologous sequence to form the D-loop structure, followed by repair DNA synthesis and resolution of recombination intermediates (18, 19, 24). During HR, RPA prevents the formation of DNA secondary structures and protects 3′-ssDNA from nucleolytic degradation (25). In addition, recent work implies a role of RPA in homology recognition (26).RPA is composed of three subunits, Rfa1, Rfa2, and Rfa3, and with a total of six oligonucleotide-binding (OB) motifs that mediate interactions with ssDNA or proteins (14, 17, 27). RPA can associate with ssDNA in different modes (28). It binds short DNA (8 to 10 nt) in an unstable mode and longer ssDNA (28 to 30 nt) in a high-affinity mode (2831). Recent single-molecule studies revealed that RPA binding on ssDNA is highly dynamic (28, 32). It can rapidly diffuse within the bound DNA ligand and quickly exchange between the free and ssDNA-bound states (3235). The cellular functions of RPA rely on its high ssDNA-binding affinity and its ability to interact with different proteins (28). Although RPA has a high affinity for ssDNA, recent studies have suggested that the binding of RPA on chromatin requires additional regulations (36). How RPA is regulated to ensure replication and repair fidelity remains poorly understood.Rtt105, a protein initially identified as a regulator of the Ty1 retrotransposon, has recently been shown to interact with RPA and acts as an RPA chaperone (36). It facilitates the nuclear localization of RPA and stimulates the loading of RPA at replication forks in unperturbed conditions or under replication stresses (36). Rtt105 exhibits synthetic genetic interactions with genes encoding replisome proteins and is required for heterochromatin silencing and telomere maintenance (37). The deletion of RTT105 results in increased gross chromosomal rearrangements and reduced resistance to DNA-damaging agents (36, 38). In vitro, Rtt105 can directly stimulate RPA binding to ssDNA, likely by changing the binding mode of RPA (36).In this study, by using a combination of genetic, biochemical, and single-molecule approaches, we demonstrate that Rtt105-dependent regulation of RPA promotes high-fidelity genome duplication and recombination while suppressing mutations and the low-fidelity repair pathways. We provide evidence that human hRIP-α, the putative functional homolog of yeast Rtt105, could regulate human RPA assembly on ssDNA in vitro. Our study unveils a layer of regulation on the maintenance of genome integrity that relies on dynamic RPA binding on ssDNA to ensure high-fidelity replication or recombination.  相似文献   

13.
Understanding molecular mechanisms in the context of living cells requires the development of new methods of in vivo biochemical analysis to complement established in vitro biochemistry. A critically important molecular mechanism is genetic recombination, required for the beneficial reassortment of genetic information and for DNA double-strand break repair (DSBR). Central to recombination is the RecA (Rad51) protein that assembles into a spiral filament on DNA and mediates genetic exchange. Here we have developed a method that combines chromatin immunoprecipitation with next-generation sequencing (ChIP-Seq) and mathematical modeling to quantify RecA protein binding during the active repair of a single DSB in the chromosome of Escherichia coli. We have used quantitative genomic analysis to infer the key in vivo molecular parameters governing RecA loading by the helicase/nuclease RecBCD at recombination hot-spots, known as Chi. Our genomic analysis has also revealed that DSBR at the lacZ locus causes a second RecBCD-mediated DSBR event to occur in the terminus region of the chromosome, over 1 Mb away.DNA double-strand break repair (DSBR) is essential for cell survival and repair-deficient cells are highly sensitive to chromosome breakage. In Escherichia coli, a single unrepaired DNA DSB per replication cycle is lethal, illustrating the critical nature of the repair reaction (1). DSBR in E. coli is mediated by homologous recombination, which relies on the RecA protein to efficiently recognize DNA sequence identity between two molecules. RecA homologs are widely conserved from bacteriophages to mammals, where they are known as the Rad51 proteins (2). The RecA protein plays its central role by binding single-stranded DNA (ssDNA) to form a presynaptic filament that searches for a homologous double-stranded DNA (dsDNA) donor from which to repair. It then catalyzes a strand-exchange reaction to form a joint molecule (3), which is stabilized by the branch migration activities of the RecG and RuvAB proteins (4). The joint molecule is then resolved by cleavage at its four-way Holliday junction by the nuclease activity of RuvABC (5, 6).RecA binding at the site of a DSB is dependent on the activity of the RecBCD enzyme (Fig. 1A). RecBCD is a helicase-nuclease that binds to dsDNA ends, then separates and unwinds the two DNA strands using the helicase activities of the RecB and RecD subunits (see refs. 7 and 8 for recent reviews). RecD is the faster motor of the two and this consequently results in the formation of a ssDNA loop ahead of RecB (Loop 1 in Fig. 1A) (9). As the enzyme translocates along dsDNA, the 3′-terminated strand is continually passed through the Chi-scanning site thought to be located in the RecC protein (10). When a Chi sequence (the octamer 5′-GCTGGTGG-3′) enters this recognition domain, the RecD motor is disengaged and the 3′ strand continues to be unwound by RecB. Under in vitro conditions, where the concentration of magnesium exceeds that of ATP, the 3′ end (unwound by RecB) is rapidly digested before Chi recognition, whereas the 5′ end (unwound by RecD) is intermittently cleaved (11, 12). After Chi recognition the 3′ end is no longer cleaved but the nuclease domain of RecB continues to degrade the 5′ end as it exits the enzyme (11, 12). Under in vitro conditions where the concentration of ATP exceeds that of magnesium, unwinding takes place but the only site of cleavage detected is ∼5 nucleotides 3′ of the Chi sequence (13, 14). Because the RecB motor continues to operate while the RecD motor is disengaged, Loop 1 is converted to a second loop located between the RecB and RecC subunits or to a tail upon release of the Chi sequence from its recognition site. We therefore describe this single-stranded region as Loop/Tail 2 in Fig. 1A. After the whole of Loop 1 is converted to Loop/Tail 2, this second single-stranded region continues to grow as long as the RecB subunit unwinds the dsDNA. The RecBCD enzyme enables RecA protein to load on to Loop/Tail 2 to generate the presynaptic filament necessary to search for homology and initiate strand-exchange (15). Finally, the RecBCD enzyme stops translocation and disassembles as it dissociates from the DNA, releasing a DNA-free RecC subunit (16).Open in a separate windowFig. 1.DSBR in E. coli. (A and B) Schematic representation of DSB processing by the RecBCD complex. (A) Before Chi recognition, both the RecB and RecD motors progress along the DNA. RecD is the faster motor and as a result a loop of ssDNA (Loop 1) is formed ahead of the slower RecB motor. The 3′ ssDNA strand is scanned for the Chi sequence by the RecC protein. (B) After Chi recognition, RecBCD likely undergoes a conformational change so that only the RecB motor is engaged. The RecA protein is recruited by the RecB nuclease domain and loaded onto the ssDNA loop generated by RecB unwinding to promote RecA nucleoprotein filament formation. In this schematic representation, the Chi site is shown held in its recognition site. However, the Chi site will be released either by disassembly of the RecBCD complex or at some point before this and the second single-stranded region on the 3′ terminating strand will be converted from a loop to a tail. Therefore, this region is denoted Loop/Tail 2. The mathematical model described in SI Appendix does not depend on the ATP/magnesium dependent differential cleavage of DNA strands (7, 8), nor does it depend on the precise time that the 3′ end is released from the complex following Chi recognition. (C) The hairpin endonuclease SbcCD is used to cleave a 246-bp interrupted palindrome inserted in the lacZ gene of the E. coli chromosome. Cleavage of this DNA hairpin results in the generation of a site-specific DSB on only one of a pair of replicating sister chromosomes, thus leaving an intact sister chromosome to serve as a template for repair by homologous recombination.Our understanding of the action of RecBCD and RecA has been the result of more than 40 years of genetic analysis and biochemical investigation of these purified proteins in vitro. However, relatively little is known about their activities on the genomic scale. To investigate these reactions in vivo, we have used RecA chromatin immunoprecipitation with next-generation sequencing (ChIP-Seq) in an experimental system that allows us to introduce a single and fully repairable DSB into the chromosome of E. coli (1). Because DSBR by homologous recombination normally involves the repair of a broken chromosome by copying the information on an unbroken sister chromosome, our laboratory has previously developed a procedure for the cleavage of only one copy of two genetically identical sister chromosomes (1). We have made use of the observation that the hairpin nuclease SbcCD specifically cleaves only one of the two sister chromosomes following DNA replication through a 246-bp interrupted palindrome to generate a two-ended DSB (1). As shown in Fig. 1B, this break is fully repairable and we have shown that recombination-proficient cells suffer very little loss of fitness in repairing such breaks (17).Here we investigate in vivo and in a quantitative manner the first steps of DSBR: because the outcome of RecBCD action is understood to be the loading of RecA on DNA in a Chi-dependent manner, we use RecA-ChIP to reveal the consequences of RecBCD action on a genomic scale during DSBR. Analyses of most ChIP-Seq datasets focus on the identification of regions of significant enrichment of a given protein but do not take into account the underlying mechanisms giving rise to the binding (18). We reasoned that given the detailed mechanistic understanding of RecBCD in vitro, we could gain a deeper insight into its in vivo functions by developing a mathematical model of RecBCD action that would enable us to estimate the mechanistic parameters of the complex in live cells. Our ChIP data indicate that RecA is indeed loaded on to DNA in a Chi-dependent manner and we have used our mathematical model to infer the parameters of RecBCD action in vivo on a genomic scale. Furthermore, our analysis reveals that DSBR at lacZ induces DSBR in the terminus region of the chromosome, an unanticipated observation illuminated by the genomic scale of our data.  相似文献   

14.
Canonical chromosomal translocations juxtaposing antigen receptor genes and oncogenes are a hallmark of many lymphoid malignancies. These translocations frequently form through the joining of DNA ends from double-strand breaks (DSBs) generated by the recombinase activating gene (RAG)-1 and -2 proteins at lymphocyte antigen receptor loci and breakpoint targets near oncogenes. Our understanding of chromosomal breakpoint target selection comes primarily from the analyses of these lesions, which are selected based on their transforming properties. RAG DSBs are rarely resolved aberrantly in wild-type developing lymphocytes. However, in ataxia telangiectasia mutated (ATM)-deficient lymphocytes, RAG breaks are frequently joined aberrantly, forming chromosomal lesions such as translocations that predispose (ATM)-deficient mice and humans to the development of lymphoid malignancies. Here, an approach that minimizes selection biases is used to isolate a large cohort of breakpoint targets of aberrantly resolved RAG DSBs in Atm-deficient lymphocytes. Analyses of this cohort revealed that frequently, the breakpoint targets for aberrantly resolved RAG breaks are other DSBs. Moreover, these nonselected lesions exhibit a bias for using breakpoints in cis, forming small chromosomal deletions, rather than breakpoints in trans, forming chromosomal translocations.  相似文献   

15.
The Mre11-Rad50-Nbs1 complex (MRN) is important for repairing DNA double-strand breaks (DSBs) by homologous recombination (HR). The endonuclease activity of MRN is critical for resecting 5′-ended DNA strands at DSB ends, producing 3′-ended single-strand DNA, a prerequisite for HR. This endonuclease activity is stimulated by Ctp1, the Schizosaccharomyces pombe homolog of human CtIP. Here, with purified proteins, we show that Ctp1 phosphorylation stimulates MRN endonuclease activity by inducing the association of Ctp1 with Nbs1. The highly conserved extreme C terminus of Ctp1 is indispensable for MRN activation. Importantly, a polypeptide composed of the conserved 15 amino acids at the C terminus of Ctp1 (CT15) is sufficient to stimulate Mre11 endonuclease activity. Furthermore, the CT15 equivalent from CtIP can stimulate human MRE11 endonuclease activity, arguing for the generality of this stimulatory mechanism. Thus, we propose that Nbs1-mediated recruitment of CT15 plays a pivotal role in the activation of the Mre11 endonuclease by Ctp1/CtIP.

DNA double-strand breaks (DSBs) are potentially lethal lesions that threaten genomic integrity and cell viability. DSBs can occur spontaneously as a result of faulty DNA metabolism or by exposure to genotoxins. In eukaryotes, these DSBs have “dirty ends” that lack ligatable 3ʹ-hydroxyl/5ʹ-phosphate groups and are often firmly attached to proteins such as the Ku70-80 heterodimer and topoisomerases (1, 2). During meiosis, the topoisomerase-like Spo11 protein generates DSBs and remains covalently attached to the 5ʹ DNA ends (3). To enable further processing, these DSB ends must be converted to “clean” ends with 3ʹ-hydroxyl/5ʹ-phosphate groups properly exposed. This step is achieved by endonucleolytic cleavage, or clipping, by Mre11 (47).In mammals, the MRE11, RAD50, and NBS1 complex (Mre11-Rad50-Nbs1 [MRN]), together with CtIP, is involved in the clipping reaction. MRE11 is the nuclease subunit that has both endonuclease and 3′-to-5′ exonuclease activities, but only the former is essential for clipping (4, 812). RAD50, a member of the Structural Maintenance of Chromosomes protein family, binds to MRE11 to form an (MRE11)2-(RAD50)2 ring structure (MR complex) (1315). NBS1 binds to the MR complex via MRE11 to form the MRN complex (16). Homologs of CtIP include Ctp1 in Schizosaccharomyces pombe and Sae2 in Saccharomyces cerevisiae (1719). Upon phosphorylation, these proteins physically interact with their cognate MRN complex via the N-terminal forkhead-associated domain of NBS1, leading to activation of the MRE11 endonucleolytic clipping activity (2024). However, the mechanistic details underlying this activation have not yet been determined.Through biochemical reconstitution using fission yeast proteins, we made three key findings regarding how Ctp1 activates MRN. First, MRN activation is mediated by Ctp1 phosphorylation, which promotes the direct association of Ctp1 with the Nbs1 subunit of MRN. Second, the highly conserved extreme C terminus of Ctp1 retains the ability to promote the endonuclease activity of MRN. Strikingly, a synthetic polypeptide comprising the 15 amino acids from the extreme C terminus of Ctp1 was sufficient for the full activation of MRN. Third, we verified the evolutionary significance of these findings by demonstrating that the conserved C-terminal polypeptide of CtIP can also stimulate the endonuclease activity of human MRN. Together, our results strongly suggest that the Ctp1-promoted MRN activation mechanism consists of at least two fundamentally separable elements: phosphorylation-induced Ctp1-MRN association and activation of MRN by the C-terminal peptide of Ctp1. Thus, recruitment of the Ctp1 C terminus to MRN is likely pivotal in this activation mechanism.  相似文献   

16.
Akt is a robust oncogene that plays key roles in the development and progression of many cancers, including glioma. We evaluated the differential propensities of the Akt isoforms toward progression in the well-characterized RCAS/Ntv-a mouse model of PDGFB-driven low grade glioma. A constitutively active myristoylated form of Akt1 did not induce high-grade glioma (HGG). In stark contrast, Akt2 and Akt3 showed strong progression potential with 78% and 97% of tumors diagnosed as HGG, respectively. We further revealed that significant variations in polarity and hydropathy values among the Akt isoforms in both the pleckstrin homology domain (P domain) and regulatory domain (R domain) were critical in mediating glioma progression. Gene expression profiles from representative Akt-derived tumors indicated dominant and distinct roles for Akt3, consisting primarily of DNA repair pathways. TCGA data from human GBM closely reflected the DNA repair function, as Akt3 was significantly correlated with a 76-gene signature DNA repair panel. Consistently, compared with Akt1 and Akt2 overexpression models, Akt3-expressing human GBM cells had enhanced activation of DNA repair proteins, leading to increased DNA repair and subsequent resistance to radiation and temozolomide. Given the wide range of Akt3-amplified cancers, Akt3 may represent a key resistance factor.Akt is among the most hyperactivated signaling pathways in human cancer and is an important kinase that regulates key cellular functions including cell growth, proliferation, angiogenesis, glucose metabolism, invasion, and survival, among others (13). Analysis of human glioblastomas (GBMs) from The Cancer Genome Atlas (TCGA) has revealed that the Akt signaling pathway is one of the top altered pathways in GBM (4, 5). Additionally, researchers have observed that the Akt activation status correlates with glioma grade (6). However, much less is known regarding the precise roles of the Akt isoforms (Akt1, Akt2, Akt3) in this disease.Akt1, Akt2, and Akt3 share roughly 80% overall sequence identity and each contain three similar domains: a pleckstrin homology, kinase, and regulatory domain (7). Although they appear to be very similar proteins, developmental studies as well as assessment of the isoforms in cancer models have revealed that they possess distinct functions (1, 8). The most extensive work on isoform differences has been performed in breast cancer. Akt2 is reported to promote migration, invasion, and metastasis, whereas Akt1 is inhibitory (8, 9). Specific phosphorylation of palladin, an actin bundling protein, by Akt1 led to decreased migration (10). Conversely, Akt2 has been found to promote breast cancer epithelial to mesenchymal transition (EMT) through miR-200 modulation (11) as well as an interaction with Snail1 on the E-cadherin promoter (12). Functional differences in glioma have been slightly variable by using knockdown systems in transformed cells. The first studies were reported on Akt2 only, which was found to mediate invasion in rat C6 glioma cells (13), and inhibition of Akt2 prolonged survival in an orthotopic rat model (14). Subsequent reports have found that Akt2, and possibly Akt3, are important for disease progression and maintenance, whereas Akt1 does not play a major role (15, 16). Another study in transformed mouse astrocytes indicate that Akt isoform function is potentially modulated by the genetic landscape of the tumor (17). The underlying basis behind these differences and the major pathways that the isoforms use remains largely unknown. To our knowledge, our study is the first to use a glial-specific transgenic mouse model to demonstrate Akt3 as a powerful inducer of glioma progression and to further reveal the previously unidentified DNA repair function of Akt3.  相似文献   

17.
18.
Interstrand cross‐links (ICLs) are extremely toxic DNA lesions that prevent DNA double‐helix separation due to the irreversible covalent linkage binding of some agents on DNA strands. Agents that induce these ICLs are thus widely used as chemotherapeutic drugs but may also lead to tumor growth. Fanconi anemia (FA) is a rare genetic disorder that leads to ICL sensitivity. This review provides update on current understanding of the role of FA proteins in repairing ICLs at various stages of cell cycle. We also discuss link between DNA cross‐link genotoxicity caused by aldehydes in FA pathway. Besides this, we summarize various ICL agents that act as drugs to treat different types of tumors and highlight strategies for modulating ICL sensitivity for therapeutic interventions that may be helpful in controlling cancer and life‐threatening disease, FA.  相似文献   

19.
Male and female germ cells can transmit genetic defects that lead to pregnancy loss, infant mortality, birth defects, and genetic diseases in offspring; however, the parental origins of transmitted defects are not random, with de novo mutations and chromosomal structural aberrations transmitted predominantly by sperm. We tested the hypotheses that paternal mutagenic exposure during late spermatogenesis can induce damage that persists in the fertilizing sperm and that the risk of embryos with paternally transmitted chromosomal aberrations depends on the efficiency of maternal DNA repair during the first cycle after fertilization. We show that female mice with defective DNA double-strand break repair had significantly increased frequencies of zygotes with sperm-derived chromosomal aberrations after matings with wild-type males irradiated 7 days earlier with 4 Gy of ionizing radiation. These findings demonstrate that mutagenic exposures during late spermatogenesis can induce damage that persists for at least 7 days in the fertilizing sperm and that maternal genotype plays a major role in determining the risks for pregnancy loss and frequencies of offspring with chromosomal defects of paternal origin.  相似文献   

20.
Given its significant role in the maintenance of genomic stability, histone methylation has been postulated to regulate DNA repair. Histone methylation mediates localization of 53BP1 to a DNA double-strand break (DSB) during homologous recombination repair, but a role in DSB repair by nonhomologous end-joining (NHEJ) has not been defined. By screening for histone methylation after DSB induction by ionizing radiation we found that generation of dimethyl histone H3 lysine 36 (H3K36me2) was the major event. Using a novel human cell system that rapidly generates a single defined DSB in the vast majority of cells, we found that the DNA repair protein Metnase (also SETMAR), which has a SET histone methylase domain, localized to an induced DSB and directly mediated the formation of H3K36me2 near the induced DSB. This dimethylation of H3K36 improved the association of early DNA repair components, including NBS1 and Ku70, with the induced DSB, and enhanced DSB repair. In addition, expression of JHDM1a (an H3K36me2 demethylase) or histone H3 in which K36 was mutated to A36 or R36 to prevent H3K36me2 formation decreased the association of early NHEJ repair components with an induced DSB and decreased DSB repair. Thus, these experiments define a histone methylation event that enhances DNA DSB repair by NHEJ.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号