FANCM, BRCA1, and BLM Cooperatively Resolve the Replication Stress at the ALT Telomeres

Total Page:16

File Type:pdf, Size:1020Kb

FANCM, BRCA1, and BLM Cooperatively Resolve the Replication Stress at the ALT Telomeres FANCM, BRCA1, and BLM cooperatively resolve the replication stress at the ALT telomeres Xiaolei Pana, William C. Drosopoulosb, Louisa Sethia, Advaitha Madireddyb, Carl L. Schildkrautb, and Dong Zhanga,1 aDepartment of Biomedical Science, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY 11568; and bDepartment of Cell Biology, Albert Einstein College of Medicine, New York, NY 10461 Edited by James E. Haber, Brandeis University, Waltham, MA, and approved June 13, 2017 (received for review May 16, 2017) In the mammalian genome, certain genomic loci/regions pose greater cells have to overcome the telomere dysfunction-induced cell cycle challenges to the DNA replication machinery (i.e., the replisome) than arrest or cell death by adopting one of the two telomere mainte- others. Such known genomic loci/regions include centromeres, com- nance mechanism (TMMs): (i)85–90% of cancers reactivate the mon fragile sites, subtelomeres, and telomeres. However, the detailed expression of telomerase (8), and (ii) the remaining 10–15% of mechanism of how mammalian cells cope with the replication stress at cancers use the HR-based ALT pathway (9, 10). these loci/regions is largely unknown. Here we show that depletion of The unique sequence and structure of mammalian telomeres FANCM, or of one of its obligatory binding partners, FAAP24, MHF1, render them especially challenging to the replisome. First, one of and MHF2, induces replication stress primarily at the telomeres of cells the strands of telomere is rich in guanines (thus called the G-rich that use the alternative lengthening of telomeres (ALT) pathway as strand) and is prone to the formation of G4s (11, 12). Second, the their telomere maintenance mechanism. Using the telomere-specific subtelomeric/telomeric noncoding RNA, TERRA, was shown to single-molecule analysis of replicated DNA technique, we found that form an R-loop (13, 14). Third, we showed previously that DNA depletion of FANCM dramatically reduces the replication efficiency at replication can be initiated from the telomeres; however, the ALT telomeres. We further show that FANCM, BRCA1, and BLM are majority of the DNA replication near the end of a mammalian actively recruited to the ALT telomeres that are experiencing replication chromosome is initiated from the subtelomeres (15), suggesting stress and that the recruitment of BRCA1 and BLM to these damaged that the subtelomeric/telomeric regions lack DNA replication or- telomeres is interdependent and is regulated by both ATR and Chk1. igins. In addition, de Lange and colleagues observed that low Mechanistically, we demonstrated that, in FANCM-depleted ALT cells, dosage of aphidicolin, a DNA polymerase inhibitor, dramatically BRCA1 and BLM help to resolve the telomeric replication stress by increases the incidence of fragile telomeres (16). Together, these stimulating DNA end resection and homologous recombination (HR). two features indicate that telomeres may be a special type of CFS. Consistent with their roles in resolving the replication stress induced by It is also known that ALT cells have more heterogeneous telo- FANCM deficiency, simultaneous depletion of BLM and FANCM, or of meres and that some of them can be quite long (17). Therefore, BRCA1 and FANCM, leads to increased micronuclei formation and synthetic lethality in ALT cells. We propose that these synthetic lethal ALT telomeres may be even more prone to express the fragility. interactions can be explored for targeting the ALT cancers. Fourth, heterochromatins are enriched at the telomeric and sub- telomeric regions of the genome (18). Finally, to protect the end – FANCM | BRCA1 | BLM | replication stress | ALT telomeres of linear chromosomes, there are a variety of high-order DNA DNA and DNA–protein structures at the telomeres. For example, the telomeric G-strand overhang folds back, invades the internal aithfully replicating its genome is vital for the fitness and double-stranded regions of telomere, and forms the telomeric health of a mammalian cell. The replisome frequently en- F loop (T-loop) as well as the displacement-loop (D-loop) (19, 20). counters a variety of impediments throughout the genome. The The T-loop and D-loop are further stabilized by the Shelterin temporary/transient slowing or stalling of the replication fork is complex and other proteins (21, 22). These various high-order referred to as replication stress (1, 2). The list of endogenous sources of replication stress is still growing. Nonetheless, the well- structures may also slow down the progression of the replisome. recognized sources of replication stress include unrepaired DNA lesions, mis-incorporated ribonucleotides, unique DNA sequences Significance that are prone to form secondary structures (e.g., G-quadruplex or G4), collision of the replication fork with the transcriptional ma- Currently, chemotherapy is still the only treatment option for chinery, an RNA–DNA hybrid (or R-loop) that is formed between ALT cancers. ALT cells rely heavily on homologous recombination a nascent RNA and the adjacent displaced single-stranded DNA to maintain their telomere length and are more susceptible to (ssDNA), common fragile sites (CFS), and tightly packed genomic replication stress. A better understanding of how ALT cells re- regions, such as heterochromatin (2). Because of these constant spond to replication stress at their telomeres will help to uncover challenges faced by the replisome, mammalian cells have developed novel therapeutic strategies to treat ALT cancers. Here we found elaborate and complex strategies to resolve the replication stress that depletion of FANCM or its obligatory binding partners in- and ensure the successful completion of DNA replication (1–4). duces pronounced replication stress at ALT telomeres. We thus One of the endogenous loci/regions that frequently pose chal- propose that FANCM-depleted ALT cells can be used as an en- lenges to the replisome is the telomere. Mammalian telomeres dogenous replication stress model. Most importantly, we pro- are tandem repetitive DNA sequences [the large majority as pose that the new synthetic lethal interactions discovered in our study can be explored for targeting ALT cancers. (TTAGGG)n] located at the end of every linear chromosome. The addition of TTAGGG is catalyzed by an enzyme called telomerase, Author contributions: X.P., W.C.D., L.S., A.M., C.L.S., and D.Z. designed research; X.P., a large ribonucleoprotein complex with reverse transcriptase activ- W.C.D., L.S., A.M., and D.Z. performed research; X.P., W.C.D., L.S., A.M., C.L.S., and D.Z. ity. The length of mammalian telomeres varies from 10 to 15 kb analyzed data; and X.P., C.L.S., and D.Z. wrote the paper. in humans and 25–50 kb in mice (5). Because most somatic cells The authors declare no conflict of interest. do not express telomerase, they experience progressive telomere This article is a PNAS Direct Submission. shortening with each cell cycle, which eventually leads to cellular 1To whom correspondence should be addressed. Email: [email protected]. senescence and aging of the organism (6). One of the hallmarks of This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. cancers is their replicative immortality (7). To achieve that, cancer 1073/pnas.1708065114/-/DCSupplemental. E5940–E5949 | PNAS | Published online July 3, 2017 www.pnas.org/cgi/doi/10.1073/pnas.1708065114 Downloaded by guest on October 4, 2021 Taken together, mammalian telomeres pose constant challenges single-molecule analysis of replicated DNA (SMARD) tech- PNAS PLUS to the replisome, and replication stress often takes place at the nique, we found that depletion of FANCM dramatically reduced telomeres, especially in ALT cells (16, 23, 24). Therefore, a robust the replication efficiency at ALT telomeres. We therefore propose replication stress response is vital for the successful replication that depletion of FANCM in ALT cells can be used as an en- of telomeres. dogenous replication stress model. Mechanistically, we found that, FANCM is an evolutionarily conserved ATP-dependent DNA in this endogenous replication stress model, BLM and BRCA1, helicase/translocase (25, 26). It belongs to the Fanconi anemia two of the most important HR proteins, are actively recruited to (FA) family of genes, the most recognized function of which is to the replication stress sites and help to resolve the replication stress repair the interstrand cross-linking (ICL) lesions (27, 28). FANCM by stimulating DNA end resection and promoting HR. Finally, we and its three obligatory binding partners, FAAP24, MHF1, and found that codepletion of FANCM and BLM or of FANCM and MHF2, are part of the FA core complex, which catalyzes the BRCA1 dramatically increases the micronuclei formation and monoubiquitination of FANCD2 and FANCI (ID2) (29–33). In induces synthetic lethality in ALT cells. addition, FANCM can also promote checkpoint activation and replication traversal across the ICL lesion (34–37). Furthermore, Results FANCM also plays a critical role in replication stress response. For Depletion of FANCM Induces Replication Stress at ALT Telomeres. example, in cells treated with hydroxyurea (HU), FANCM binds Based on the potential roles of FANCM in DNA replication, the checkpoint protein HCLK2 and promotes the activation of we hypothesized that the DNA damages seen in FANCM- ATR-mediated checkpoint response (34). FANCM also helps to deficient cells are due to replication stress (34, 38). Replication stabilize
Recommended publications
  • Regulation of DNA Cross-Link Repair by the Fanconi Anemia/BRCA Pathway
    Downloaded from genesdev.cshlp.org on September 29, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW Regulation of DNA cross-link repair by the Fanconi anemia/BRCA pathway Hyungjin Kim and Alan D. D’Andrea1 Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA The maintenance of genome stability is critical for sur- and quadradials, a phenotype widely used as a diagnostic vival, and its failure is often associated with tumorigen- test for FA. esis. The Fanconi anemia (FA) pathway is essential for At least 15 FA gene products constitute a common the repair of DNA interstrand cross-links (ICLs), and a DNA repair pathway, the FA pathway, which resolves germline defect in the pathway results in FA, a cancer ICLs encountered during replication (Fig. 1A). Specifi- predisposition syndrome driven by genome instability. cally, eight FA proteins (FANCA/B/C/E/F/G/L/M) form Central to this pathway is the monoubiquitination of a multisubunit ubiquitin E3 ligase complex, the FA core FANCD2, which coordinates multiple DNA repair activ- complex, which activates the monoubiquitination of ities required for the resolution of ICLs. Recent studies FANCD2 and FANCI after genotoxic stress or in S phase have demonstrated how the FA pathway coordinates three (Wang 2007). The FANCM subunit initiates the pathway critical DNA repair processes, including nucleolytic in- (Fig. 1B). It forms a heterodimeric complex with FAAP24 cision, translesion DNA synthesis (TLS), and homologous (FA-associated protein 24 kDa), and the complex resem- recombination (HR). Here, we review recent advances in bles an XPF–ERCC1 structure-specific endonuclease pair our understanding of the downstream ICL repair steps (Ciccia et al.
    [Show full text]
  • An Overview of DNA Repair Mechanisms in Plants Compared to Mammals
    Cell. Mol. Life Sci. (2017) 74:1693–1709 DOI 10.1007/s00018-016-2436-2 Cellular and Molecular Life Sciences REVIEW Protecting DNA from errors and damage: an overview of DNA repair mechanisms in plants compared to mammals Claudia P. Spampinato1 Received: 8 August 2016 / Revised: 1 December 2016 / Accepted: 5 December 2016 / Published online: 20 December 2016 Ó Springer International Publishing 2016 Abstract The genome integrity of all organisms is con- methods and reporter systems for gene expression and stantly threatened by replication errors and DNA damage function studies. Here, I provide a current understanding of arising from endogenous and exogenous sources. Such base DNA repair genes in plants, with a special focus on A. pair anomalies must be accurately repaired to prevent thaliana. It is expected that this review will be a valuable mutagenesis and/or lethality. Thus, it is not surprising that resource for future functional studies in the DNA repair cells have evolved multiple and partially overlapping DNA field, both in plants and animals. repair pathways to correct specific types of DNA errors and lesions. Great progress in unraveling these repair mecha- Keywords DNA repair Á Photolyases Á BER Á NER Á nisms at the molecular level has been made by several MMR Á HR Á NHEJ talented researchers, among them Tomas Lindahl, Aziz Sancar, and Paul Modrich, all three Nobel laureates in Chemistry for 2015. Much of this knowledge comes from Introduction studies performed in bacteria, yeast, and mammals and has impacted research in plant systems. Two plant features All organisms are constantly exposed to environmental should be mentioned.
    [Show full text]
  • Bloom Syndrome Complex Promotes FANCM Recruitment to Stalled Replication Forks and Facilitates Both Repair and Traverse of DNA Interstrand Crosslinks
    OPEN Citation: Cell Discovery (2016) 2, 16047; doi:10.1038/celldisc.2016.47 ARTICLE www.nature.com/celldisc Bloom syndrome complex promotes FANCM recruitment to stalled replication forks and facilitates both repair and traverse of DNA interstrand crosslinks Chen Ling1, Jing Huang2,4, Zhijiang Yan1, Yongjiang Li1, Mioko Ohzeki3, Masamichi Ishiai3, Dongyi Xu1,5, Minoru Takata3, Michael Seidman2, Weidong Wang1 1Lab of Genetics, National Institute on Aging, National Institute of Health, Baltimore, MD, USA; 2Lab of Molecular Gerontology, National Institute on Aging, National Institute of Health, Baltimore, MD, USA; 3Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan The recruitment of FANCM, a conserved DNA translocase and key component of several DNA repair protein complexes, to replication forks stalled by DNA interstrand crosslinks (ICLs) is a step upstream of the Fanconi anemia (FA) repair and replication traverse pathways of ICLs. However, detection of the FANCM recruitment has been technically challenging so that its mechanism remains exclusive. Here, we successfully observed recruitment of FANCM at stalled forks using a newly developed protocol. We report that the FANCM recruitment depends upon its intrinsic DNA trans- locase activity, and its DNA-binding partner FAAP24. Moreover, it is dependent on the replication checkpoint kinase, ATR; but is independent of the FA core and FANCD2–FANCI complexes, two essential components of the FA pathway, indicating that the FANCM recruitment occurs downstream of ATR but upstream of the FA pathway. Interestingly, the recruitment of FANCM requires its direct interaction with Bloom syndrome complex composed of BLM helicase, Topoisomerase 3α, RMI1 and RMI2; as well as the helicase activity of BLM.
    [Show full text]
  • A Structure-Specific Nucleic Acid-Binding Domain Conserved Among DNA Repair Proteins
    A structure-specific nucleic acid-binding domain conserved among DNA repair proteins Aaron C. Masona, Robert P. Rambob, Briana Greera, Michael Pritchetta, John A. Tainerb, David Cortezc, and Brandt F. Eichmana,c,1 aDepartment of Biological Sciences, Vanderbilt University, Nashville, TN 37232; bLife Sciences Division, Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and cDepartment of Biochemistry, Vanderbilt School of Medicine, Nashville, TN 37232 Edited by James M. Berger, Johns Hopkins University School of Medicine, Baltimore, MD, and approved April 17, 2014 (received for review December 30, 2013) SMARCAL1, a DNA remodeling protein fundamental to genome 1), also known as HARP (HepA-related protein), is one of several integrity during replication, is the only gene associated with the ATP-dependent motor proteins capable of fork regression and im- developmental disorder Schimke immuno-osseous dysplasia (SIOD). portant for genetic stability, including Rad54, RecQ paralogs, BLM, SMARCAL1-deficient cells show collapsed replication forks, S-phase WRN, FANCM, ZRANB3, HLTF/Rad5, T4 bacteriophage UvsW, cell cycle arrest, increased chromosomal breaks, hypersensitivity to archaeal HelQ/Hel308/Hjm, and Escherichia coli RecG (18–26). genotoxic agents, and chromosomal instability. The SMARCAL1 cat- SMARCAL1 is a distant SNF2 family member of dsDNA trans- alytic domain (SMARCAL1CD) is composed of an SNF2-type double- locating chromatin remodeling proteins (27) with a binding prefer- ence for branched DNA structures, and has been shown to catalyze stranded DNA motor ATPase fused to a HARP domain of unknown A function. The mechanisms by which SMARCAL1 and other DNA ATP-dependent regression of model replication forks (Fig. 1 ), branch migration of Holliday junctions, and reannealing of RPA- translocases repair replication forks are poorly understood, in part – because of a lack of structural information on the domains outside coated plasmids (28 30).
    [Show full text]
  • The Fanconi Anemia DNA Damage Repair Pathway in the Spotlight for Germline Predisposition to Colorectal Cancer
    European Journal of Human Genetics (2016) 24, 1501–1505 & 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved 1018-4813/16 www.nature.com/ejhg SHORT REPORT The Fanconi anemia DNA damage repair pathway in the spotlight for germline predisposition to colorectal cancer Clara Esteban-Jurado1, Sebastià Franch-Expósito1, Jenifer Muñoz1, Teresa Ocaña1, Sabela Carballal1, Maria López-Cerón1, Miriam Cuatrecasas2, Maria Vila-Casadesús3, Juan José Lozano3, Enric Serra4, Sergi Beltran4, The EPICOLON Consortium, Alejandro Brea-Fernández5, Clara Ruiz-Ponte5, Antoni Castells1, Luis Bujanda6, Pilar Garre7, Trinidad Caldés7, Joaquín Cubiella8, Francesc Balaguer1 and Sergi Castellví-Bel*,1 Colorectal cancer (CRC) is one of the most common neoplasms in the world. Fanconi anemia (FA) is a very rare genetic disease causing bone marrow failure, congenital growth abnormalities and cancer predisposition. The comprehensive FA DNA damage repair pathway requires the collaboration of 53 proteins and it is necessary to restore genome integrity by efficiently repairing damaged DNA. A link between FA genes in breast and ovarian cancer germline predisposition has been previously suggested. We selected 74 CRC patients from 40 unrelated Spanish families with strong CRC aggregation compatible with an autosomal dominant pattern of inheritance and without mutations in known hereditary CRC genes and performed germline DNA whole- exome sequencing with the aim of finding new candidate germline predisposition variants. After sequencing and data analysis, variant prioritization selected only those very rare alterations, producing a putative loss of function and located in genes with a role compatible with cancer. We detected an enrichment for variants in FA DNA damage repair pathway genes in our familial CRC cohort as 6 families carried heterozygous, rare, potentially pathogenic variants located in BRCA2/FANCD1, BRIP1/FANCJ, FANCC, FANCE and REV3L/POLZ.
    [Show full text]
  • Biochemical Activities and Genetic Functions of the Drosophila Melanogaster Fancm Helicase in Dna Repair
    BIOCHEMICAL ACTIVITIES AND GENETIC FUNCTIONS OF THE DROSOPHILA MELANOGASTER FANCM HELICASE IN DNA REPAIR Noelle-Erin F. Romero A dissertation submitted to the faculty of the University of North Carolina at Chapel Hill in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Curriculum of Genetics and Molecular Biology Chapel Hill 2016 Approved by: Steve Matson Jeff Sekelsky Dorothy Erie Robert Duronio Cyrus Vaziri © 2016 Noelle-Erin F. Romero ALL RIGHTS RESERVED ii ABSTRACT Noelle-Erin F. Romero: Biochemical activities and genetic functions of the Drosophila melanogaster Fancm helicase in DNA repair (Under the direction of Steve Matson and Jeff Sekelsky) The DNA damage response in eukaryotes involves multiple, complex, and often redundant pathways that respond to various types of DNA damage that affect one or both strands of DNA. One type of toxic DNA damage that can occur is a double-strand break (DSB). Repair of a DSB can lead to the formation of a recombination product known as a crossover (CO). Crossovers in mitotic cells can be deleterious and lead to chromosomal rearrangements or cell death. In order to limit crossing over during DSB repair, eukaryotes possess mechanisms to ensure crossovers do not occur. In this manner, several helicases function during repair of DSBs to promote accurate repair and prevent the formation of crossovers through homologous recombination. Among these helicases is the Fanconi anemia group M (FANCM) protein. FANCM is one of 17 Fanconi anemia (FA) proteins and is one of the most broadly? conserved FA proteins. FANCM and its orthologs, Mph1 and Fml1, are DNA junction-specific helicases/translocases that process homologous recombination (HR) intermediates.
    [Show full text]
  • Homologous Recombination Nonhomologous End Joining Interstrand Cross-Link Repair Nucleotide Excision Repair Mismatch Repair
    DNA repair pathways Homologous recombination Nonhomologous end joining Interstrand cross-link repair Nucleotide excision repair UV DNA crosslinkers Ionizing radiation Ionizing radiation Genotoxic chemicals Genotoxic chemicals Free radicals Free radicals Mechanical stress Mechanical stress Global genome Transcription FAAP24 ICL repair coupled repair MHF1 Lesion recognition and FANCM replicative fork convergence XPC HR23B MHF2 Transcription P XPE block PARP1 P P ATM ATM Mre11 Mre11 CSA H2A H2AX ATM Rad50 γ-H2AX γ- P P ATM Rad50 P P XPC HR23B RNA Pol I/II γ-H2AX Nbs1 Nbs1 SMC1 P H4K20me2 P P CSB Mre11 FANCM, FAAP24, MHF1, MHF2 (lesion recognition) P P Rad50 Mre11 P P Poly ubiquitin Nbs1 ATM P P FANCT, FANCL (D2-I ubiquitination) P 53BP1 Histones Rad50 Histones XPG ERCC1 histones Strand tethering FANCA, B, C, E, F, G TFIID Rap80 ATM Nbs1 Mdc1 P FAAP20, 100 XPD TFB5 P P Replicative helicase eviction XPB XPF Abraxas P P and fork approach at -1 position P P Rnf8 P BRCC36 RPA XPA Caesin Kinase2 Ku / Lig4 dependent Ku / Lig4 independent Ubc13 Homologous pathway pathway γH2A.X recombination XPC HR23B BRCA1 P MRN ATM complex Rag 1/2 Resection 5’ CtIP CMG 3’ V D J 3’ CtIP Facnoni 5’ Anemia Replicative helicase ERCC1 MRN Core UHRF1 ATM Resection Cdc45 XPF complex Complex XPG P MCM2-7 TFIID GINS XPA XPD XPB TFB5 V D J CMG BRCA2 RPA P Rad51BCD Hairpin DNA Bard1 BRCA1 XRCC2/3 ends Rad51 monomers ATR ub Rad52 Rad54 FANC1 P RFC Polδ/ε Rad51BCD P FANCD2 ub XRCC2/3 DNA PKcs and BRCA1 P Ku70 Rad52 Nucleolytic incisionand BRCA2 Ku70 RPA 5’ Ku80 unhooking by
    [Show full text]
  • AAA-Atpase FIDGETIN-LIKE 1 and Helicase FANCM Antagonize
    AAA-ATPase FIDGETIN-LIKE 1 and helicase FANCM antagonize meiotic crossovers by distinct mechanisms Chloé Girard, Liudmila Chelysheva, Sandrine Choinard, Nicole Froger, Nicolas Macaisne, A. Lehmemdi, Julien Mazel, Wayne Crismani, Raphaël Mercier To cite this version: Chloé Girard, Liudmila Chelysheva, Sandrine Choinard, Nicole Froger, Nicolas Macaisne, et al.. AAA-ATPase FIDGETIN-LIKE 1 and helicase FANCM antagonize meiotic crossovers by distinct mechanisms. PLoS Genetics, Public Library of Science, 2015, 11 (7), pp.e1005369. 10.1371/jour- nal.pgen.1005369. hal-01204200 HAL Id: hal-01204200 https://hal.archives-ouvertes.fr/hal-01204200 Submitted on 27 May 2020 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. RESEARCH ARTICLE AAA-ATPase FIDGETIN-LIKE 1 and Helicase FANCM Antagonize Meiotic Crossovers by Distinct Mechanisms Chloe Girard1,2, Liudmila Chelysheva1,2, Sandrine Choinard1,2, Nicole Froger1,2, Nicolas Macaisne1,2, Afef Lehmemdi1,2, Julien Mazel1,2, Wayne Crismani1,2*, Raphael Mercier1,2* 1 INRA, Institut Jean-Pierre Bourgin, UMR1318, ERL CNRS 3559, Saclay Plant Sciences, RD10, Versailles, France, 2 AgroParisTech, Institut Jean-Pierre Bourgin, UMR 1318, ERL CNRS 3559, Saclay Plant Sciences, RD10, Versailles, France a11111 * [email protected] (WC); [email protected] (RM) Abstract Meiotic crossovers (COs) generate genetic diversity and are critical for the correct comple- OPEN ACCESS tion of meiosis in most species.
    [Show full text]
  • Functional Comparison of XPF Missense Mutations Associated to Multiple DNA Repair Disorders
    G C A T T A C G G C A T genes Article Functional Comparison of XPF Missense Mutations Associated to Multiple DNA Repair Disorders Maria Marín 1, María José Ramírez 1,2, Miriam Aza Carmona 1,3,4, Nan Jia 5, Tomoo Ogi 5, Massimo Bogliolo 1,2,* and Jordi Surrallés 1,2,6,* 1 Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08028 Barcelona, Spain; [email protected] (M.M.); [email protected] (M.J.R.); [email protected] (M.A.C.) 2 Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain 3 Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, 28029 Madrid, Spain 4 CIBERER, ISCIII, 28029 Madrid, Spain 5 Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan/Department of Human Genetics and Molecular Biology, Graduate School of Medicine, Nagoya University, Nagoya 464-0805, Japan; [email protected] (N.J.); [email protected] (T.O.) 6 Genetics Department Institute of Biomedical Research, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain * Correspondence: [email protected] (M.B.); [email protected] (J.S.); Tel.:+34-935537376 (M.B.); +34-935868048 (J.S.) Received: 21 December 2018; Accepted: 11 January 2019; Published: 17 January 2019 Abstract: XPF endonuclease is one of the most important DNA repair proteins. Encoded by XPF/ERCC4, XPF provides the enzymatic activity of XPF-ERCC1 heterodimer, an endonuclease that incises at the 5’ side of various DNA lesions.
    [Show full text]
  • A Homozygous FANCM Mutation Underlies a Familial Case of Non-Syndromic Primary Ovarian Insufficiency
    RESEARCH ARTICLE A homozygous FANCM mutation underlies a familial case of non-syndromic primary ovarian insufficiency Baptiste Fouquet1†, Patrycja Pawlikowska2†, Sandrine Caburet3, Celine Guigon4, Marika Ma¨ kinen5, Laura Tanner5, Marja Hietala5, Kaja Urbanska6, Laura Bellutti7, Be´ range` re Legois3, Bettina Bessieres8, Alain Gougeon9, Alexandra Benachi10, Gabriel Livera7, Filippo Rosselli2, Reiner A Veitia3‡, Micheline Misrahi1‡* 1Faculte´ de Me´decine, Universite´ Paris Sud, Universite´ Paris Saclay, Hoˆpital Biceˆtre, Le Kremlin-Biceˆtre, France; 2CNRS UMR8200,Equipe labellise´e La Ligue Contre Le Cancer, Universite´ Paris Sud, Universite´ Paris Saclay, Gustave Roussy, Vilejuif, France; 3Institut Jacques Monod, Universite´ Paris Diderot, Paris, France; 4Universite´ Paris-Diderot, CNRS, UMR 8251, INSERM, U1133, Paris, France; 5Department of Clinical Genetics, Turku University Hospital, Turku, Finland; 6CNRS UMR8200, Universite´ Paris Sud, Universite´ Paris Saclay, Villejuif, France; 7UMR967 INSERM, CEA/DRF/iRCM/SCSR/LDG, Universite´ Paris Diderot, Sorbonne Paris Cite´, Universite´ Paris-Sud, Universite´ Paris-Saclay, Fontenay aux Roses, France; 8Department of Histology, Embryology and Cytogenetics, Hoˆpital Necker-enfants malades, Paris, France; 9UMR Inserm 1052, CNRS 5286, Faculte´ de Me´decine Laennec, Lyon, France; 10Department of Obstetrics and Gynaecology, AP-HP, Universite´ Paris-Sud, Universite´ Paris-Saclay, Clamart, France *For correspondence: [email protected] Abstract Primary Ovarian Insufficiency (POI) affects ~1%
    [Show full text]
  • The FANCM-BLM-TOP3A-RMI Complex Suppresses Alternative Lengthening of Telomeres (ALT)
    ARTICLE https://doi.org/10.1038/s41467-019-10180-6 OPEN The FANCM-BLM-TOP3A-RMI complex suppresses alternative lengthening of telomeres (ALT) Robert Lu1,5, Julienne J. O’Rourke2,3,5, Alexander P. Sobinoff1,5, Joshua A.M. Allen1, Christopher B. Nelson1, Christopher G. Tomlinson1, Michael Lee 1, Roger R. Reddel 4, Andrew J. Deans 2,3 & Hilda A. Pickett1 1234567890():,; The collapse of stalled replication forks is a major driver of genomic instability. Several committed mechanisms exist to resolve replication stress. These pathways are particularly pertinent at telomeres. Cancer cells that use Alternative Lengthening of Telomeres (ALT) display heightened levels of telomere-specific replication stress, and co-opt stalled replication forks as substrates for break-induced telomere synthesis. FANCM is a DNA translocase that can form independent functional interactions with the BLM-TOP3A-RMI (BTR) complex and the Fanconi anemia (FA) core complex. Here, we demonstrate that FANCM depletion provokes ALT activity, evident by increased break-induced telomere synthesis, and the induction of ALT biomarkers. FANCM-mediated attenuation of ALT requires its inherent DNA translocase activity and interaction with the BTR complex, but does not require the FA core complex, indicative of FANCM functioning to restrain excessive ALT activity by ameliorating replication stress at telomeres. Synthetic inhibition of FANCM-BTR complex formation is selectively toxic to ALT cancer cells. 1 Telomere Length Regulation Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead 2145 NSW, Australia. 2 Genome Stability Unit, St. Vincent’s Institute, 9 Princes St, Fitzroy 3065 VIC, Australia. 3 Department of Medicine (St.
    [Show full text]
  • Alternative Functions for Human FANCM at Telomeres
    MINI REVIEW published: 06 September 2019 doi: 10.3389/fmolb.2019.00084 ALTernative Functions for Human FANCM at Telomeres Beatriz Domingues-Silva, Bruno Silva and Claus M. Azzalin* Instituto de Medicina Molecular João Lobo Antunes (iMM), Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal The human FANCM ATPase/translocase is involved in various cellular pathways including DNA damage repair, replication fork remodeling and R-loop resolution. Recently, reports from three independent laboratories have disclosed a previously unappreciated role for FANCM in telomerase-negative human cancer cells that maintain their telomeres through the Alternative Lengthening of Telomeres (ALT) pathway. In ALT cells, FANCM limits telomeric replication stress and damage, and, in turn, ALT activity by suppressing accumulation of telomeric R-loops and by regulating the action of the BLM helicase. As a consequence, FANCM inactivation leads to exaggerated ALT activity and ultimately cell death. The studies reviewed here not only unveil a novel function for human FANCM, but also point to this enzyme as a promising target for anti-ALT cancer therapy. Keywords: FANCM, telomeres, ALT, R-loops, TERRA, BLM helicase FANCM Edited by: Human FANConi anemia, complementation group M (FANCM) is a highly conserved protein with Gaelle Legube, ATPase and DNA translocase activity, belonging to the Fanconi anemia (FA) core complex (Meetei FR3743 Centre de Biologie Intégrative et al., 2005). FA is a hereditary disorder characterized by bone marrow failure, hypersensitivity (CBI), France to agents inducing DNA interstrand crosslinks (ICLs), chromosomal abnormalities and, later in Reviewed by: life, cancer. Although FANCM is part of the FA complex, FANCM mutations are not causative Lee Zou, of FA (Singh et al., 2009; Bogliolo et al., 2018; Catucci et al., 2018).
    [Show full text]