scholarly journals Mispair-bound human MutS–MutL complex triggers DNA incisions and activates mismatch repair

Cell Research ◽  
2021 ◽  
Author(s):  
Janice Ortega ◽  
Grace Sanghee Lee ◽  
Liya Gu ◽  
Wei Yang ◽  
Guo-Min Li

AbstractDNA mismatch repair (MMR) relies on MutS and MutL ATPases for mismatch recognition and strand-specific nuclease recruitment to remove mispaired bases in daughter strands. However, whether the MutS–MutL complex coordinates MMR by ATP-dependent sliding on DNA or protein–protein interactions between the mismatch and strand discrimination signal is ambiguous. Using functional MMR assays and systems preventing proteins from sliding, we show that sliding of human MutSα is required not for MMR initiation, but for final mismatch removal. MutSα recruits MutLα to form a mismatch-bound complex, which initiates MMR by nicking the daughter strand 5′ to the mismatch. Exonuclease 1 (Exo1) is then recruited to the nick and conducts 5′ → 3′ excision. ATP-dependent MutSα dissociation from the mismatch is necessary for Exo1 to remove the mispaired base when the excision reaches the mismatch. Therefore, our study has resolved a long-standing puzzle, and provided new insights into the mechanism of MMR initiation and mispair removal.

DNA Repair ◽  
2016 ◽  
Vol 38 ◽  
pp. 50-57 ◽  
Author(s):  
Peter Friedhoff ◽  
Pingping Li ◽  
Julia Gotthardt

2017 ◽  
Author(s):  
Najla Al-Sweel ◽  
Vandana Raghavan ◽  
Abhishek Dutta ◽  
V. P. Ajith ◽  
Luigi Di Vietro ◽  
...  

AbstractMlh1-Mlh3 is an endonuclease hypothesized to act in meiosis to resolve double Holliday junctions into crossovers. It also plays a minor role in eukaryotic DNA mismatch repair (MMR). To understand how Mlh1-Mlh3 functions in both meiosis and MMR, we analyzed in baker’s yeast 60 new mlh3 alleles. Five alleles specifically disrupted MMR, whereas one (mlh3-32) specifically disrupted meiotic crossing over. Mlh1-mlh3 representatives for each separation of function class were purified and characterized. Both Mlh1-mlh3-32 (MMR+, crossover-) and Mlh1-mlh3-45 (MMR-, crossover+) displayed wild-type endonuclease activities in vitro. Msh2-Msh3, an MSH complex that acts with Mlh1-Mlh3 in MMR, stimulated the endonuclease activity of Mlh1-mlh3-32 but not Mlh1-mlh3-45, suggesting that Mlh1-mlh3-45 is defective in MSH interactions. Whole genome recombination maps were constructed for two mlh3 mutants with opposite separation of function phenotypes, and an endonuclease defective mutant. Unexpectedly, all three showed increases in the number of non-crossover events that were not observed in mlh3Δ. Our observations provide a structure-function map for Mlh3 that reveals the importance of protein-protein interactions in regulating Mlh1-Mlh3’s enzymatic activity. They also illustrate how defective meiotic components can alter the fate of meiotic recombination intermediates, providing new insights for how meiotic recombination pathways are regulated.Author SummaryDuring meiosis, diploid germ cells that become eggs or sperm undergo a single round of DNA replication followed by two consecutive chromosomal divisions. The segregation of chromosomes at the first meiotic division is dependent in most organisms on at least one genetic exchange, or crossover event, between chromosome homologs. Homologs that do not receive a crossover frequently undergo non-disjunction at the first meiotic division, yielding gametes that lack chromosomes or contain additional copies. Such events have been linked to human disease and infertility. Recent studies suggest that the Mlh1-Mlh3 complex is an endonuclease that resolves recombination intermediates into crossovers. Interestingly, this complex also acts as a matchmaker in DNA mismatch repair (MMR) to remove DNA replication errors. How does one complex act in two different processes? We investigated this question by performing a mutational analysis of the baker’s yeast Mlh3 protein. Five mutations were identified that disrupted MMR but not crossing over, and one mutation disrupted crossing over while maintaining MMR. Using a combination of biochemical and genetic analyses to further characterize these mutants we illustrate the importance of protein-protein interactions for Mlh1-Mlh3’s activity. Importantly, we illustrate how defective meiotic components can alter the outcome of meiotic recombination events. They also provide new insights in our understanding of the basis of infertility syndromes.


2007 ◽  
Vol 35 (8) ◽  
pp. 2609-2619 ◽  
Author(s):  
Nina Østergaard Knudsen ◽  
Finn Cilius Nielsen ◽  
Lena Vinther ◽  
Ronni Bertelsen ◽  
Steen Holten-Andersen ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2526-2526
Author(s):  
Amar Desai ◽  
Yulan Qing ◽  
Min Liu ◽  
Jonathan Kenyon ◽  
Uchenna Nwafor ◽  
...  

Abstract Abstract 2526 Poster Board II-503 The DNA mismatch repair pathway (MMR) is a fundamental process in cells that functions to correct mispaired bases and insertion/deletion loops caused by errors in replication. Failure in MMR can lead to the accumulation of mutations and carcinogenesis, notably hereditary nonpolyposis colorectal cancer (HNPCC). In hematopoiesis, loss of MMR results in methylating agent resistance and an HSC repopulation defect. Our research focused on the significance of Exonuclease 1, the enzyme responsible for excising mispaired bases in MMR. Interestingly Exo1−/− mice display a much milder pathogenic phenotype when compared to the MMR deficient MSH2−/− mice; characterized by lower mutation rates, lower levels of microsatellite instability, and in humans no association with HNPCC. These findings led us to hypothesize that the limited phenotype observed in Exo1−/− mice is due to (a) complementation through another exonuclease occurs which restores the proficiency of the MMR pathway or (b) an alternative pathway exists which allows for exonuclease independent repair. To test this hypothesis we derived primary MEFs from WT, Exo1−/− and MSH2−/− mice and performed temozolomide sensitivity assays. We observed that Exo1−/− MEFs are similarly sensitive as WT cells when treated with the drug, confirming proficient MMR, while MSH2−/− MEFs display a decreased sensitivity. Additionally, comet assays with the same cell populations show a persistence of DNA single strand breaks in the Exo1−/− and WT cells 24 hours post temozolomide treatment, consistent with proficient MMR activity, while MSH2−/− MEFs show no such damage. To confirm functional MMR in hematopoiesis of Exo1−/− mice we conducted a competitive repopulation study. We found that Exo1−/− and WT marrow engraft at similar levels 8 and 16 weeks post transplantation. We subsequently treated these mice with 80 mg/kg temozolomide to determine if Exo1−/− marrow progenitors would confer a competitive survival advantage post treatment. Consistent with our hypothesis we found that the ratio of marrow cells remained approximately 50:50 (Exo1:WT) in contrast to data obtained from MSH2−/− mice in which the MSH2−/− cells display a strong 95:5 (MSH2:WT) advantage post drug treatment. This strongly suggests functional MMR in the Exo1 deficient mice. We next coimmunoprecipitated the MMR complex by pulling down Mlh1 in Exo1−/− and WT MEFs. Exo1, a key exonuclease in mismatch repair, does not modulate hematopoietic function as do the other MMR proteins and appears to be complemented by an as yet undefined exonuclease. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Felipe A. Calil ◽  
Bin-Zhong Li ◽  
Kendall A. Torres ◽  
Katarina Nguyen ◽  
Nikki Bowen ◽  
...  

AbstractEukaryotic DNA Mismatch Repair (MMR) involves redundant exonuclease 1 (Exo1)-dependent and Exo1-independent pathways, of which the Exo1-independent pathway(s) is not well understood. The exo1Δ440-702 mutation, which deletes the MutS Homolog 2 (Msh2) and MutL Homolog 1 (Mlh1) interacting peptides (SHIP and MIP boxes, respectively), eliminates the Exo1 MMR functions but is not lethal in combination with rad27Δ mutations. Analyzing the effect of different combinations of the exo1Δ440-702 mutation, a rad27Δ mutation and the pms1-A99V mutation, which inactivates an Exo1-independent MMR pathway, demonstrated that each of these mutations inactivates a different MMR pathway. Furthermore, it was possible to reconstitute a Rad27- and Msh2-Msh6-dependent MMR reaction in vitro using a mispaired DNA substrate and other MMR proteins. Our results demonstrate Rad27 defines an Exo1-independent eukaryotic MMR pathway that is redundant with at least two other MMR pathways.


Sign in / Sign up

Export Citation Format

Share Document