DNA Double-Strand Break Formation and Repair in Tetrahymena Meiosis

Total Page:16

File Type:pdf, Size:1020Kb

DNA Double-Strand Break Formation and Repair in Tetrahymena Meiosis G Model YSCDB-1977; No. of Pages 9 ARTICLE IN PRESS Seminars in Cell & Developmental Biology xxx (2016) xxx–xxx Contents lists available at ScienceDirect Seminars in Cell & Developmental Biology journal homepage: www.elsevier.com/locate/semcdb Review DNA double-strand break formation and repair in Tetrahymena meiosis a,∗ b Josef Loidl , Alexander Lorenz a Department of Chromosome Biology, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-Gasse 9, A-1030 Vienna, Austria b Institute of Medical Sciences (IMS), University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK a r t i c l e i n f o a b s t r a c t Article history: The molecular details of meiotic recombination have been determined for a small number of model Received 2 November 2015 organisms. From these studies, a general picture has emerged that shows that most, if not all, recombina- Accepted 12 February 2016 tion is initiated by a DNA double-strand break (DSB) that is repaired in a recombinogenic process using a Available online xxx homologous DNA strand as a template. However, the details of recombination vary between organisms, and it is unknown which variant is representative of evolutionarily primordial meiosis or most preva- Keywords: lent among eukaryotes. To answer these questions and to obtain a better understanding of the range Meiosis of recombination processes among eukaryotes, it is important to study a variety of different organisms. Recombination Protist Here, the ciliate Tetrahymena thermophila is introduced as a versatile meiotic model system, which has DSB the additional bonus of having the largest phylogenetic distance to all of the eukaryotes studied to date. Crossover Studying this organism can contribute to our understanding of the conservation and diversification of meiotic recombination processes. © 2016 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). Contents 1. Introduction . 00 1.1. Meiotic recombination . 00 1.2. Studying meiosis in Tetrahymena . 00 2. Stages of recombination . 00 2.1. DSB initiation . 00 2.2. DSB end processing. .00 2.3. DNA strand exchange . 00 2.4. The timing of DSB formation and repair . 00 2.5. Crossing over . 00 2.6. Pro-crossover factors . 00 2.6.1. Hop2 . 00 2.6.2. Msh4, Msh5 . 00 2.6.3. Sgs1 . 00 2.7. Alternative DSB repair pathways . 00 3. Crossover control . 00 3.1. The question of CO interference . 00 3.2. Tetrahymena crossing over does not conform to the two-pathway paradigm . 00 3.3. Why is Tetrahymena meiosis “simple”? . 00 4. Conclusions and outlook . 00 Acknowledgements. .00 References . 00 ∗ Corresponding author. E-mail addresses: [email protected] (J. Loidl), [email protected] (A. Lorenz). http://dx.doi.org/10.1016/j.semcdb.2016.02.021 1084-9521/© 2016 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). Please cite this article in press as: J. Loidl, A. Lorenz, DNA double-strand break formation and repair in Tetrahymena meiosis, Semin Cell Dev Biol (2016), http://dx.doi.org/10.1016/j.semcdb.2016.02.021 G Model YSCDB-1977; No. of Pages 9 ARTICLE IN PRESS 2 J. Loidl, A. Lorenz / Seminars in Cell & Developmental Biology xxx (2016) xxx–xxx 1. Introduction passed on to sexual progeny. The polyploid macronucleus repre- sents the soma. Protein-encoding genes are only transcribed from 1.1. Meiotic recombination the macronucleus. It disintegrates in sexual cells and is reconsti- tuted in progeny cells from new micronuclei. Meiosis is a specialized cell division in which germ progenitor Tetrahymena has many benefits as an experimental model sys- cells reduce the somatic double chromosome set to a gametic single tem for molecular and genetic studies [20]. It is easy to culture, and set during sexual reproduction in eukaryotes. The chromosomes of thanks to its inducible and highly synchronous meiosis, Tetrahy- both original sets are randomly represented in the gametic set. In mena is the only organism besides the yeasts, where bulk isolation addition to this mixing of original sets, the chromosomes of the of meiotic DNA for molecular assays of DSBs is possible. Its nuclei gametic set are mosaics that are assembled from portions of the and chromosomes are amenable to cytological analysis. Addition- homologous parental chromosomes. This “interhomolog recom- ally, due to its capability of performing both vegetative propagation bination” provides the physical linkage of chromosome pairs and and sexual reproduction, meiosis-defective mutants can be easily ensures their faithful segregation. Moreover, it contributes to the maintained [21]. reshuffling of parental genes in the gametes and the genetic diver- Mating pairs undergo simultaneous meioses [22], and the pro- sity of the offspring. gression of meiosis can easily be followed and staged. Perhaps the Each chromosome consists of two sister DNA molecules, the most remarkable feature of Tetrahymena meiosis is the extreme chromatids. At certain locations, one of the four chromatids of a elongation of nuclei during prophase to approximately twice the chromosome pair experiences a programmed DNA double-strand length of the cell (Fig. 2). Within an elongated nucleus, chro- break (DSB) that is caused by a dedicated transesterase, Spo11 mosomes are arranged in a polarized manner, resembling the (Fig. 1) (see Ref. [1]). This DSB is repaired using one of the sister- widely-conserved bouquet where all telomeres gather in a limited or homologous DNA strands as a template for repair synthesis. The area at the nuclear periphery [23]. However, in Tetrahymena, in repair process may lead to the exchange of strands in a subset of addition to telomere clustering at one end of the nucleus, the cen- events. During meiosis, several regulatory mechanisms ensure that tromeres are attached to the opposite end. This ultimate bouquet this exchange takes place more often with a homologous chromatid arrangement promotes the juxtaposition of homologous regions than a sister chromatid (see Ref. [2] for a detailed review on these and, thereby, homologous pairing and crossing over [24,25]. Cyto- steps of recombination). logical studies failed to detect a SC [25–27]. Following this unusual DNA flanking the DSB is resected to expose single-strand (ss) 3 pairing stage, nuclei shorten and five condensed bivalents become overhangs. These ssDNA tracts are first bound by proteins that sta- discernible at the diplotene/diakinesis stage (Fig. 2), which is fol- bilize and reconfigure the DNA. Later, these proteins are replaced lowed by first and second meiotic divisions within the intact by the strand exchange proteins Rad51 and/or Dmc1. These DNA- nuclear envelope. protein complexes can invade homologous dsDNA tracts, search for BLAST searches revealed homologs of several conserved mei- complementary base sequences and initiate strand.
Recommended publications
  • SMX Makes the Cut in Genome Stability
    www.impactjournals.com/oncotarget/ Oncotarget, 2017, Vol. 8, (No. 61), pp: 102765-102766 Editorial SMX makes the cut in genome stability Haley D.M. Wyatt and Stephen C. West The faithful duplication and preservation of our SLX4 scaffold co-ordinate three different nucleases for genetic material is essential for cell survival; however, DNA cleavage? SMX was found to be a promiscuous DNA is susceptible to damage by extracellular and endonuclease that cleaves a broad range of DNA secondary intracellular agents (e.g. ultraviolet radiation, reactive structures in vitro3. Remarkably, SLX4 co-ordinates the oxygen species). DNA double-strand breaks (DSBs) SLX1 and MUS81-EME1 nucleases during HJ resolution are thought to represent the most dangerous type of [3, 4] (Figure 1). The involvement of two active sites from lesion, as the failure to repair a DSB can lead to loss of different heterodimeric enzymes leads to a non-canonical genetic information, chromosomal rearrangements, and mechanism of HJ resolution. It was also shown that SLX4 cell death. Fortunately, cells contain sophisticated DNA activates MUS81-EME1 to cleave structures that resemble repair pathways to counteract the deleterious effects of stalled replication forks [3] (Figure 1). Activation involves genotoxic agents. Mutations in DNA repair genes are relaxation of MUS81-EME1’s substrate specificity, which linked to various diseases, including neurological defects, is regulated by a helix-hairpin-helix (HhH) domain in immunodeficiency, and cancer. the MUS81 N-terminus (MUS81 N-HhH). Intriguingly, Gross chromosomal rearrangements, including MUS81 N-HhH also mediates the interaction with deletions, duplications, and translocations, are a hallmark SLX4 via a C-terminal SAP domain (SLX4 SAP) [5].
    [Show full text]
  • The Choice in Meiosis – Defining the Factors That Influence Crossover Or Non-Crossover Formation
    Commentary 501 The choice in meiosis – defining the factors that influence crossover or non-crossover formation Jillian L. Youds and Simon J. Boulton* DNA Damage Response Laboratory, Cancer Research UK, London Research Institute, Clare Hall, Blanche Lane, South Mimms EN6 3LD, UK *Author for correspondence ([email protected]) Journal of Cell Science 124, 501-513 © 2011. Published by The Company of Biologists Ltd doi:10.1242/jcs.074427 Summary Meiotic crossovers are essential for ensuring correct chromosome segregation as well as for creating new combinations of alleles for natural selection to take place. During meiosis, excess meiotic double-strand breaks (DSBs) are generated; a subset of these breaks are repaired to form crossovers, whereas the remainder are repaired as non-crossovers. What determines where meiotic DSBs are created and whether a crossover or non-crossover will be formed at any particular DSB remains largely unclear. Nevertheless, several recent papers have revealed important insights into the factors that control the decision between crossover and non-crossover formation in meiosis, including DNA elements that determine the positioning of meiotic DSBs, and the generation and processing of recombination intermediates. In this review, we focus on the factors that influence DSB positioning, the proteins required for the formation of recombination intermediates and how the processing of these structures generates either a crossover or non-crossover in various organisms. A discussion of crossover interference, assurance and homeostasis, which influence crossing over on a chromosome-wide and genome-wide scale – in addition to current models for the generation of interference – is also included. This Commentary aims to highlight recent advances in our understanding of the factors that promote or prevent meiotic crossing over.
    [Show full text]
  • Cleavage Mechanism of Human Mus81–Eme1 Acting on Holliday-Junction Structures
    Cleavage mechanism of human Mus81–Eme1 acting on Holliday-junction structures Ewan R. Taylor* and Clare H. McGowan*†‡ Departments of *Molecular Biology and †Cell Biology, The Scripps Research Institute, La Jolla, CA 92037 Edited by Stephen J. Elledge, Harvard Medical School, Boston, MA, and approved January 10, 2008 (received for review October 29, 2007) Recombination-mediated repair plays a central role in maintaining Results genomic integrity during DNA replication. The human Mus81–Eme1 Recombinant Human Mus81–Eme1. Truncation analysis of both endonuclease is involved in recombination repair, but the exact Mus81 and Eme1 was used to define the minimal domains structures it acts on in vivo are not known. Using kinetic and enzy- required for endonuclease function [see supplementary infor- matic analysis of highly purified recombinant enzyme, we find mation (SI) Fig. 5]. Versions of each protein containing amino that Mus81–Eme1 catalyzes coordinate bilateral cleavage of acids 260–551 for Mus81 and amino acids 244–571 for Eme1 model Holliday-junction structures. Using a self-limiting, cruciform- were expressed in Escherichia coli. The recombinant truncated containing substrate, we demonstrate that bilateral cleavage occurs complex, which we named EcME, was well expressed, largely sequentially within the lifetime of the enzyme–substrate complex. soluble, and, as detailed below, active. The complex was purified Coordinate bilateral cleavage is promoted by the highly cooperative to apparent homogeneity through affinity chromatography, ion nature of the enzyme and results in symmetrical cleavage of a exchange, and gel filtration steps (Fig. 1a). cruciform structure, thus, Mus81–Eme1 can ensure coordinate, bilat- eral cleavage of Holliday junction-like structures.
    [Show full text]
  • Regulation of Mus81-Eme1 Structure-Specific Endonuclease by Eme1 SUMO-Binding and Rad3atr Kinase Is Essential in the Absence of Rqh1blm Helicase
    bioRxiv preprint doi: https://doi.org/10.1101/2021.07.29.454171; this version posted July 29, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Regulation of Mus81-Eme1 structure-specific endonuclease by Eme1 SUMO-binding and Rad3ATR kinase is essential in the absence of Rqh1BLM helicase Giaccherini C.1,#, Scaglione S. 1, Coulon S. 1, Dehé P.M.1,#,* and Gaillard P.H.L. 1* 1 Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm, CNRS, Aix- Marseille Université, Institut Paoli-Calmettes, Marseille, France. #The authors contributed equally *correspondence [email protected] [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.07.29.454171; this version posted July 29, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract The Mus81-Eme1 structure-specific endonuclease is crucial for the processing of DNA recombination and late replication intermediates. In fission yeast, stimulation of Mus81-Eme1 in response to DNA damage at the G2/M transition relies on Cdc2CDK1 and DNA damage checkpoint-dependent phosphorylation of Eme1 and is critical for chromosome stability in absence of the Rqh1BLM helicase. Here we identify Rad3ATR checkpoint kinase consensus phosphorylation sites and two SUMO interacting motifs (SIM) within a short N-terminal domain of Eme1 that is required for cell survival in absence of Rqh1BLM.
    [Show full text]
  • The MUS81 Endonuclease Is Essential for Telomerase Negative Cell Proliferation Sicong Zeng Washington University School of Medicine in St
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2009 The MUS81 endonuclease is essential for telomerase negative cell proliferation Sicong Zeng Washington University School of Medicine in St. Louis Qin Yang Washington University School of Medicine in St. Louis Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Zeng, Sicong and Yang, Qin, ,"The UM S81 endonuclease is essential for telomerase negative cell proliferation." Cell Cycle.8,15. 2157-2160. (2009). https://digitalcommons.wustl.edu/open_access_pubs/3006 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. [Cell Cycle 8:15, 2157-2160; 1 August 2009]; ©2009 Landes Bioscience Extra View The MUS81 endonuclease is essential for telomerase negative cell proliferation Sicong Zeng† and Qin Yang* Department of Radiation Oncology; Washington University School of Medicine; St. Louis, MO USA †Present address: Institute of Reproduction and Stem Cell Engineering; Central South University; Changsha, Hunan China Key words: ALT, APBs, MUS81, telomere recombination A substantial number of human tumors (~10%) are telomerase breaks and maintains telomere length and stability. Progressive negative, and cells in such tumors have been proposed to main- telomere shortening in normal cells during DNA replication leads tain telomere length by the alternative lengthening of telomeres eventually to a permanent halt of cell division referred to as replica- (ALT) pathway. Although details of the molecular mechanism tive senescence. This end replication problem of human telomeres of ALT are largely unknown, previous studies have shown that has received particular attention due to its implications in ageing telomere homologous recombination (HR) is implicated in the and cancer.
    [Show full text]
  • Structure of the Germline Genome of Tetrahymena Thermophila
    1 Structure of the germline genome of Tetrahymena thermophila 2 and relationship to the massively rearranged somatic genome 3 4 Eileen P. Hamiltona#, Aurélie Kapustab#, Piroska E. Huvosc, Shelby L. Bidwelld, Nikhat Zafard, 5 Haibao Tangd, Michalis Hadjithomasd, Vivek Krishnakumard, Jonathan Badgerd, Elisabet V. 6 Calerd, Carsten Russe, Qiandong Zenge, Lin Fane, Joshua Z. Levine, Terrance Sheae, Sarah K. 7 Younge, Ryan Hegartye, Riza Dazae, Sharvari Gujjae, Jennifer R. Wortmane, Bruce Birrene, 8 Chad Nusbaume, Jainy Thomasb, Clayton M. Careyb, Ellen J. Prithamb, Cédric Feschotteb, 9 Tomoko Notof, Kazufumi Mochizukif, Romeo Papazyang, Sean D. Tavernag, Paul H. Dearh, 10 Donna M. Cassidy-Hanleyi, Jie Xiongj, Wei Miaoj, Eduardo Oriasa, Robert S. Coyned* 11 12 a: Department of Molecular, Cellular, and Developmental Biology, University of California, 13 Santa Barbara, CA, USA 14 b: Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 15 USA 16 c: Department of Medical Biochemistry, Southern Illinois University, Carbondale, IL, USA. 17 d: J. Craig Venter Institute, Rockville, MD, USA 18 e: Eli and Edythe L. Broad Institute of Harvard and MIT, Cambridge, MA, USA 19 f: Institute of Molecular Biotechnology, Vienna, Austria; current address: Institute of 20 Human Genetics - CNRS, Montpellier, France 21 g: Department of Pharmacology and Molecular Sciences, The Johns Hopkins University 22 School of Medicine, Baltimore, MD, USA 1 23 h: MRC Laboratory of Molecular Biology, Cambridge, UK 24 i: Dept. of Microbiology and Immunology, Cornell University Veterinary Medical Center, 25 Ithaca, NY, USA 26 j: Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, People's Republic of 27 China 28 *: corresponding author.
    [Show full text]
  • Roles of SLX1–SLX4, MUS81–EME1, and GEN1 in Avoiding Genome Instability and Mitotic Catastrophe
    Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press Roles of SLX1–SLX4, MUS81–EME1, and GEN1 in avoiding genome instability and mitotic catastrophe Shriparna Sarbajna,1 Derek Davies,2 and Stephen C. West1,3 1Clare Hall Laboratories, Cancer Research UK, London Research Institute, Herts EN6 3LD, United Kingdom; 2London Research Institute, London WC2A 3PX, United Kingdom The resolution of recombination intermediates containing Holliday junctions (HJs) is critical for genome maintenance and proper chromosome segregation. Three pathways for HJ processing exist in human cells and involve the following enzymes/complexes: BLM–TopoIIIa–RMI1–RMI2 (BTR complex), SLX1–SLX4–MUS81– EME1 (SLX–MUS complex), and GEN1. Cycling cells preferentially use the BTR complex for the removal of double HJs in S phase, with SLX–MUS and GEN1 acting at temporally distinct phases of the cell cycle. Cells lacking SLX–MUS and GEN1 exhibit chromosome missegregation, micronucleus formation, and elevated levels of 53BP1-positive G1 nuclear bodies, suggesting that defects in chromosome segregation lead to the transmission of extensive DNA damage to daughter cells. In addition, however, we found that the effects of SLX4, MUS81, and GEN1 depletion extend beyond mitosis, since genome instability is observed throughout all phases of the cell cycle. This is exemplified in the form of impaired replication fork movement and S-phase progression, endogenous checkpoint activation, chromosome segmentation, and multinucleation. In contrast to SLX4, SLX1, the nuclease subunit of the SLX1–SLX4 structure-selective nuclease, plays no role in the replication-related phenotypes associated with SLX4/MUS81 and GEN1 depletion.
    [Show full text]
  • Msh4 and Msh5 Function in SC-Independent Chiasma Formation During the Streamlined Meiosis of Tetrahymena
    HIGHLIGHTED ARTICLE INVESTIGATION Msh4 and Msh5 Function in SC-Independent Chiasma Formation During the Streamlined Meiosis of Tetrahymena Anura Shodhan,* Agnieszka Lukaszewicz,* Maria Novatchkova,†,‡ and Josef Loidl*,1 *Department of Chromosome Biology and Max F. Perutz Laboratories, Center for Molecular Biology, University of Vienna, A-1030 Vienna, Austria, †Research Institute of Molecular Pathology, A-130 Vienna, Austria, and ‡Institute of Molecular Biotechnology of the Austrian Academy of Sciences, A-1030 Vienna, Austria ORCID ID: 0000-0002-3675-2146 (A.S.) ABSTRACT ZMM proteins have been defined in budding yeast as factors that are collectively involved in the formation of interfering crossovers (COs) and synaptonemal complexes (SCs), and they are a hallmark of the predominant meiotic recombination pathway of most organisms. In addition to this so-called class I CO pathway, a minority of crossovers are formed by a class II pathway, which involves the Mus81-Mms4 endonuclease complex. This is the only CO pathway in the SC-less meiosis of the fission yeast. ZMM proteins (including SC components) were always found to be co-occurring and hence have been regarded as functionally linked. Like the fission yeast, the protist Tetrahymena thermophila does not possess a SC, and its COs are dependent on Mus81-Mms4. Here we show that the ZMM proteins Msh4 and Msh5 are required for normal chiasma formation, and we propose that they have a pro-CO function outside a canonical class I pathway in Tetrahymena. Thus, the two-pathway model is not tenable as a general rule. EIOSIS is a specialized cell division by which the dip- half in budding yeast (Mancera et al.
    [Show full text]
  • ATR Protects the Genome Against R Loops Through a MUS81-Triggered Feedback Loop
    ATR Protects the Genome against R loops through a MUS81-Triggered Feedback Loop The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Matos, Dominick Amaral. 2020. ATR Protects the Genome against R loops through a MUS81-Triggered Feedback Loop. Doctoral dissertation, Harvard University, Graduate School of Arts & Sciences. Citable link https://nrs.harvard.edu/URN-3:HUL.INSTREPOS:37365155 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA ATR Protects the Genome against R loops through a MUS81-Triggered Feedback Loop A dissertation presented by Dominick A Matos to The Division of Medical Sciences in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the subject of Biological and Biomedical Sciences Harvard University Cambridge, Massachusetts December 2019 @ 2019 Dominick A Matos All rights reserved. Dissertation Advisor: Lee Zou Dominick A Matos ATR Protects the Genome against R loops through a MUS81-Triggered Feedback Loop Abstract R loops are three-stranded transcription intermediates consisting of RNA:DNA hybrids and displaced single-stranded DNA. Although R loop formation is important for a number of cellular processes, they can cause genomic instability if dysregulated. The R loop-induced genomic instability is largely dependent on DNA replication, suggesting that it may arise from the collisions between R loops and DNA replication forks. How cells respond to the collisions between R loops and replication fork is poorly understood.
    [Show full text]
  • Two Distinct MUS81-EME1 Complexes from Arabidopsis Thaliana Process
    Two Distinct MUS81-EME1 Complexes from Arabidopsis Process Holliday Junctions1[W] Verena Geuting2, Daniela Kobbe, Frank Hartung, Jasmin Du¨ rr3, Manfred Focke, and Holger Puchta* Botanik II, Universita¨t Karlsruhe, 76128 Karlsruhe, Germany The MUS81 endonuclease complex has been shown to play an important role in the repair of stalled or blocked replication forks and in the processing of meiotic recombination intermediates from yeast to humans. This endonuclease is composed of two subunits, MUS81 and EME1. Surprisingly, unlike other organisms, Arabidopsis (Arabidopsis thaliana) has two EME1 homologs encoded in its genome. AtEME1A and AtEME1B show 63% identity on the protein level. We were able to demonstrate that, after expression in Escherichia coli, each EME1 protein can assemble with the unique AtMUS81 to form a functional endonuclease. Both complexes, AtMUS81-AtEME1A and AtMUS81-AtEME1B, are not only able to cleave 3#-flap structures and nicked Holliday junctions (HJs) but also, with reduced efficiency, intact HJs. While the complexes have the same cleavage patterns with both nicked DNA substrates, slight differences in the processing of intact HJs can be detected. Our results are in line with an involvement of both MUS81-EME1 endonuclease complexes in DNA recombination and repair processes in Arabidopsis. DNA is constantly damaged by extrinsic and intrin- nucleases and forms a functional endonuclease com- sic factors, such as UV and ionizing irradiation, chem- plex with its interaction partner EME1 (also referred to icals, and cellular processes. The accumulation of as MMS4 in Saccharomyces cerevisiae; Boddy et al., 2000; DNA damage can impair essential processes, includ- Interthal and Heyer, 2000; Chen et al., 2001).
    [Show full text]
  • Bi-Allelic MCM10 Variants Associated with Immune Dysfunction and Cardiomyopathy Cause Telomere Shortening
    ARTICLE https://doi.org/10.1038/s41467-021-21878-x OPEN Bi-allelic MCM10 variants associated with immune dysfunction and cardiomyopathy cause telomere shortening Ryan M. Baxley 1, Wendy Leung1,11, Megan M. Schmit 1,11, Jacob Peter Matson 2,11, Lulu Yin1, Marissa K. Oram 1, Liangjun Wang1, John Taylor3, Jack Hedberg1, Colette B. Rogers1, Adam J. Harvey1, Debashree Basu1, Jenny C. Taylor4,5, Alistair T. Pagnamenta 4,5, Helene Dreau6, Jude Craft7, Elizabeth Ormondroyd 8, Hugh Watkins 8, Eric A. Hendrickson 1, Emily M. Mace 9, Jordan S. Orange9, ✉ Hideki Aihara 1, Grant S. Stewart 10, Edward Blair7, Jeanette Gowen Cook 2 & Anja-Katrin Bielinsky 1 1234567890():,; Minichromosome maintenance protein 10 (MCM10) is essential for eukaryotic DNA repli- cation. Here, we describe compound heterozygous MCM10 variants in patients with dis- tinctive, but overlapping, clinical phenotypes: natural killer (NK) cell deficiency (NKD) and restrictive cardiomyopathy (RCM) with hypoplasia of the spleen and thymus. To understand the mechanism of MCM10-associated disease, we modeled these variants in human cell lines. MCM10 deficiency causes chronic replication stress that reduces cell viability due to increased genomic instability and telomere erosion. Our data suggest that loss of MCM10 function constrains telomerase activity by accumulating abnormal replication fork structures enriched with single-stranded DNA. Terminally-arrested replication forks in MCM10- deficient cells require endonucleolytic processing by MUS81, as MCM10:MUS81 double mutants display decreased viability and accelerated telomere shortening. We propose that these bi-allelic variants in MCM10 predispose specific cardiac and immune cell lineages to prematurely arrest during differentiation, causing the clinical phenotypes observed in both NKD and RCM patients.
    [Show full text]
  • Supporting Information
    Supporting Information Cnossen et al. 10.1073/pnas.1309438111 SI Text gs.washington.edu/EVS/ (in 6,500 individuals variants were Investigations in Polycystic Liver Disease-1 Family. Ultrasonography. assessed in the project) Ultrasound images of liver and kidneys were acquired using Database of Single Nucleotide Polymorphisms (dbSNP): www. a 3.6-MHz general purpose clinical echo system (Acuson ×150; ncbi.nlm.nih.gov/projects/SNP/; Bethesda (MD): National Cen- Siemens AG) equipped with a curved linear array transducer in ter for Biotechnology Information, National Library of Medi- all 39 members of proband III/18. Presence or absence of hepatic cine; NCBI dbSNP Build 137; 26th June 2012 available and/or renal cysts was investigated and noted carefully by at least Mouse Genome Informatics; v.MGI 5.12 (last database up- two clinicians (W.R.C., M.C., and J.P.H.D.). These ultrasound date 03–13-2013): www.informatics.jax.org/searches/allele_ examinations were conducted in all 39 members within 2 wk. report.cgi?_Marker_key=37359;MGD During the project, five members (II/13; III/13; III/14; III/21; and Human gene mutation database (HGMD Professional) (www. III/26) were reevaluated by abdominal ultrasonography (Table biobase-international.com/hgmd) from BIOBASE Corpora- S4). Member III/22 could not undergo a second evaluation due tion; Professional 2012.4, 14 December 2012 accessed to chronic health issues. A complete clinical analysis of the liver MRS database; v.6 (latest version): http://mrs.cmbi.ru.nl/m6/ and kidney phenotype in these individuals by CT or MRI scan- entry?db=sprot&id=lrp5_human&rq=lrp5_human ning was not ethically approved for this project.
    [Show full text]