ZCCHC8, the Nuclear Exosome Targeting Component, Is Mutated in Familial Pulmonary Fibrosis and Is Required for Telomerase RNA Maturation

Total Page:16

File Type:pdf, Size:1020Kb

ZCCHC8, the Nuclear Exosome Targeting Component, Is Mutated in Familial Pulmonary Fibrosis and Is Required for Telomerase RNA Maturation Downloaded from genesdev.cshlp.org on October 7, 2021 - Published by Cold Spring Harbor Laboratory Press ZCCHC8, the nuclear exosome targeting component, is mutated in familial pulmonary fibrosis and is required for telomerase RNA maturation Dustin L. Gable,1,2,3 Valeriya Gaysinskaya,2,3 Christine C. Atik,2,3 C. Conover Talbot Jr.,4 Byunghak Kang,5 Susan E. Stanley,1,2,3 Elizabeth W. Pugh,6 Nuria Amat-Codina,2,3 Kara M. Schenk,7 Murat O. Arcasoy,8 Cory Brayton,5 Liliana Florea,6 and Mary Armanios2,3,6,9,10 1Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; 2Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA; 3Telomere Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA; 4Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; 5Department of Comparative and Molecular Pathobiology, 6Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA; 7Osler Medical Housestaff Training Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; 8Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27708, USA; 9Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA Short telomere syndromes manifest as familial idiopathic pulmonary fibrosis; they are the most common premature aging disorders. We used genome-wide linkage to identify heterozygous loss of function of ZCCHC8, a zinc-knuckle containing protein, as a cause of autosomal dominant pulmonary fibrosis. ZCCHC8 associated with TR and was required for telomerase function. In ZCCHC8 knockout cells and in mutation carriers, genomically extended tel- omerase RNA (TR) accumulated at the expense of mature TR, consistent with a role for ZCCHC8 in mediating TR 3′ end targeting to the nuclear RNA exosome. We generated Zcchc8-null mice and found that heterozygotes, similar to −/− human mutation carriers, had TR insufficiency but an otherwise preserved transcriptome. In contrast, Zcchc8 −/− mice developed progressive and fatal neurodevelopmental pathology with features of a ciliopathy. The Zcchc8 brain transcriptome was highly dysregulated, showing accumulation and 3′ end misprocessing of other low-abun- dance RNAs, including those encoding cilia components as well as the intronless replication-dependent histones. Our data identify a novel cause of human short telomere syndromes-familial pulmonary fibrosis and uncover nuclear exosome targeting as an essential 3′ end maturation mechanism that vertebrate TR shares with replication-de- pendent histones. [Keywords: lung disease; ciliopathy; nuclear RNA exosome; RNA processing; telomerase RNA] Supplemental material is available for this article. Received April 19, 2019; revised version accepted August 15, 2019. Telomerase, the specialized ribonucleoprotein that syn- Armanios 2012; McNally et al. 2019). Among IPF families thesizes telomere DNA, has two essential components: with known mutations, half carry mutations in TERT or the telomerase reverse transcriptase, TERT, and, a spe- TR (Armanios et al. 2007; Tsakiri et al. 2007). The rest car- cialized non-coding RNA, TR (also known as TERC), ry mutations in one of five other telomere maintenance which provides the template for telomere repeat addition genes, RTEL1, PARN, NAF1, TINF2, and DKC1 (Alder (Greider and Blackburn 1985, 1987, 1989; Feng et al. 1995; et al. 2013, 2015; Cogan et al. 2015; Stuart et al. Lingner et al. 1997). Mutant telomerase genes have their 2015; Stanley et al. 2016). Four of these seven known mu- most common manifestation in the age-related disease id- tant telomere genes in familial IPF affect TR itself, its iopathic pulmonary fibrosis (IPF) (Armanios et al. 2007; deadenylation, trafficking, or stability as in the case for PARN, NAF1, and DKC1, respectively (Mitchell et al. 1999b; Moon et al. 2015; Stanley et al. 2016). However, 10Present address: Department of Medicine, Massachusetts General Hos- for 50%–60% of IPF families, the genetic etiology is pital, Boston, MA 02114, USA. Corresponding author: [email protected] Article published online ahead of print. Article and publication date are © 2019 Gable et al. This article, published in Genes & Development,is online at http://www.genesdev.org/cgi/doi/10.1101/gad.326785.119. Free- available under a Creative Commons License (Attribution-NonCommer- ly available online through the Genes & Development Open Access cial 4.0 International), as described at http://creativecommons.org/licens- option. es/by-nc/4.0/. GENES & DEVELOPMENT 33:1–16 Published by Cold Spring Harbor Laboratory Press; ISSN 0890-9369/19; www.genesdev.org 1 Downloaded from genesdev.cshlp.org on October 7, 2021 - Published by Cold Spring Harbor Laboratory Press Gable et al. unknown, and within this subset there are individuals This led us to determine preclinical status by measuring with unexplained TR insufficiency and short telomere TR levels in LCLs and we found the proband’s two chil- syndrome features (Stanley et al. 2016). dren also had 50% of control TR levels, while the remain- Here, we used an unbiased genome-wide linkage ap- ing relatives had TR levels similar to healthy controls proach to discover a novel familial IPF disease gene that (Fig. 1B). Telomere length measurement confirmed both is required for TR maturation and telomerase function. of these “low TR” asymptomatic individuals were at Human TR has its own RNA polymerase II transcriptional risk for disease since they had abnormally short telomere unit (Feng et al. 1995); it shares a 3′ end box H/ACA motif length, below the first age-adjusted percentile, and shorter with a subset of non-coding RNAs, but, relative to them, it than the proband (Fig. 1C). These data supported assigning has low abundance (Mitchell et al. 1999a; Chen et al. affected status to the two presymptomatic individuals, 2000; Stanley et al. 2016). Telomere maintenance is vul- and the other family members as unaffected (Fig. 1D). nerable to small reductions in TR levels (Greider 2006; We used these designations to perform a SNP array-based McNally et al. 2019), and TR is haploinsufficient in both genome-wide linkage analysis and identified a single humans and mice (Hathcock et al. 2002; Hao et al. 2005; 17.3-Mb linkage peak on chromosome 12 (1,073,260– Armanios et al. 2009). TR+/− mice show genetic anticipa- 1,246,120) with a maximum log of the odds ratio (LOD) tion with later generations eventually developing short score of 2.99 (Fig. 1E). Whole-genome sequencing identi- telomere phenotypes including bone marrow failure and fied three rare coding variants within this interval in the immunodeficiency (Hathcock et al. 2002; Hao et al. proband (Supplemental Table S1), but only one of them 2005; Armanios et al. 2009; Wagner et al. 2018). The deter- fell in an RNA-related gene, ZCCHC8. The variant, minants of vertebrate TR integrity are incompletely un- c.C557T predicted a p.P186L missense, had not been re- derstood in part because the TR sequence is divergent, ported in >140,000 individuals in the databases (Supple- and different organisms have adapted distinct posttran- mental Table S1) and segregated with both the IPF and scriptional processing strategies (Chen et al. 2000; Podlev- low TR phenotypes (Fig. 1D). In a multi-species align- sky and Chen 2016). Tetrahymena thermophila TR, for ment, p.P186 fell in an unstructured, highly conserved example, is transcribed by RNA polymerase III and relies domain of ZCCHC8 (Fig. 1F; Supplemental Fig. S1). We on runs of T’s for transcriptional termination (Greider and screened 42 other genetically uncharacterized families Blackburn 1989). S. pombe, on the other hand, uses a mod- with pulmonary fibrosis and other short telomere syn- ified spliceosome-dependent cleavage mechanism as an drome phenotypes but did not identify additional initial 3′ end processing step (Box et al. 2008; Coy et al. ZCCHC8 variants, suggesting these mutations are rare 2013). Vertebrate TR is transcribed as a longer RNA (1 of 43, 2%), and consistent with a locus heterogeneity (Feng et al. 1995; Goldfarb and Cech 2013), and a subset for short telomere syndromes. of these extended forms, the short extended oligoadeny- lated ones, were recently found to be precursors for the ZCCHC8P186L is a loss-of-function mutation mature functional TR (Roake et al. 2019). While TR’s deg- radation has been linked to the RNA exosome in some We first tested the functional effects of ZCCHC8P186L and cell-based systems (Nguyen et al. 2015; Tseng et al. found that all three mutation carriers had 50% lower 2015), the mechanisms by which a longer extended TR ZCCHC8 protein levels compared with controls, includ- matures into a shorter, functional RNA are not known. ing noncarrier relatives (LCLs; P = 0.038, Mann-Whitney We report here mutant ZCCHC8, the zinc finger U-test) (Fig. 2A; antibody validation in Supplemental CCHC-type domain containing 8 protein, and a compo- Fig. S2A). Low protein levels were also seen in the pro- nent of the nuclear exosome targeting complex (NEXT) band’s primary skin fibroblasts and were specific to (Lubas et al. 2011), is a cause of familial IPF. We show ZCCHC8 as the levels of the other two NEXT complex that ZCCHC8 is required for TR maturation and uncover proteins, RBM7 and SKIV2L2, were normal (Fig. 2B; two distinct RNA dysregulation
Recommended publications
  • Large-Scale Analysis of Genome and Transcriptome Alterations in Multiple Tumors Unveils Novel Cancer-Relevant Splicing Networks
    Downloaded from genome.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Research Large-scale analysis of genome and transcriptome alterations in multiple tumors unveils novel cancer-relevant splicing networks Endre Sebestyén,1,5 Babita Singh,1,5 Belén Miñana,1,2 Amadís Pagès,1 Francesca Mateo,3 Miguel Angel Pujana,3 Juan Valcárcel,1,2,4 and Eduardo Eyras1,4 1Universitat Pompeu Fabra, E08003 Barcelona, Spain; 2Centre for Genomic Regulation, E08003 Barcelona, Spain; 3Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), E08908 L’Hospitalet del Llobregat, Spain; 4Catalan Institution for Research and Advanced Studies, E08010 Barcelona, Spain Alternative splicing is regulated by multiple RNA-binding proteins and influences the expression of most eukaryotic genes. However, the role of this process in human disease, and particularly in cancer, is only starting to be unveiled. We system- atically analyzed mutation, copy number, and gene expression patterns of 1348 RNA-binding protein (RBP) genes in 11 solid tumor types, together with alternative splicing changes in these tumors and the enrichment of binding motifs in the alter- natively spliced sequences. Our comprehensive study reveals widespread alterations in the expression of RBP genes, as well as novel mutations and copy number variations in association with multiple alternative splicing changes in cancer drivers and oncogenic pathways. Remarkably, the altered splicing patterns in several tumor types recapitulate those of undifferen- tiated cells. These patterns are predicted to be mainly controlled by MBNL1 and involve multiple cancer drivers, including the mitotic gene NUMA1. We show that NUMA1 alternative splicing induces enhanced cell proliferation and centrosome am- plification in nontumorigenic mammary epithelial cells.
    [Show full text]
  • Comprehensive Analysis of Differentially Expressed Lncrnas Mirnas and Mrna and Their Cerna Network of Patients with Rare-Earth Pneumoconiosis
    fgene-12-700398 July 13, 2021 Time: 17:18 # 1 ORIGINAL RESEARCH published: 19 July 2021 doi: 10.3389/fgene.2021.700398 Comprehensive Analysis of Differentially Expressed lncRNAs miRNAs and mRNA and Their ceRNA Network of Patients With Rare-Earth Pneumoconiosis Xue-min Shi, Yu-chao Bai, Yan-rong Gao, Ning Bu, Hai-yan Song, Li-hua Huang, Yu-hang Zhao* and Su-hua Wang* School of Public Health, Baotou Medical College, Baotou, China Rare-earth pneumoconiosis (REP) is the main occupational disease of rare earth exposed workers and there is no specific treatment. In this study, we performed high-throughput sequencing on the plasma of nine REP to describe and analyze the expression profiles of long non-coding RNA (lncRNA), micro RNA (miRNA) and Edited by: Duc-Hau Le, mRNA and investigate their regulatory networks. Our results identified a total of 125 Vingroup Big Data Institute, Vietnam lncRNAs, 5 miRNAs, and 82 mRNAs were differentially expressed in the plasma of Reviewed by: patients with REP. Furthermore, Ontology (GO) and Kyoto Encyclopedia of Genes and Eman Toraih, Genomes (KEGG) analysis were used to analyze the differentially expressed non-coding Tulane University, United States Isha Monga, RNAs (ncRNA). We found the differential expression of ncRNA are mainly related to Columbia University Irving Medical the response of cells to stimulation, Hedgehog signaling pathway and so on. We Center, United States also constructed lncRNA-miRNA-mRNA networks to further explore their underlying *Correspondence: Yu-hang Zhao mechanism and possible relationships in REP. We found that in the competitive [email protected] endogenous RNA (ceRNA) networks, lncRNA acts as a sponge of miRNA to regulate the Su-hua Wang target gene.
    [Show full text]
  • Proteomic and Metabolomic Analyses of Mitochondrial Complex I-Deficient
    THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 287, NO. 24, pp. 20652–20663, June 8, 2012 © 2012 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. Proteomic and Metabolomic Analyses of Mitochondrial Complex I-deficient Mouse Model Generated by Spontaneous B2 Short Interspersed Nuclear Element (SINE) Insertion into NADH Dehydrogenase (Ubiquinone) Fe-S Protein 4 (Ndufs4) Gene*□S Received for publication, November 25, 2011, and in revised form, April 5, 2012 Published, JBC Papers in Press, April 25, 2012, DOI 10.1074/jbc.M111.327601 Dillon W. Leong,a1 Jasper C. Komen,b1 Chelsee A. Hewitt,a Estelle Arnaud,c Matthew McKenzie,d Belinda Phipson,e Melanie Bahlo,e,f Adrienne Laskowski,b Sarah A. Kinkel,a,g,h Gayle M. Davey,g William R. Heath,g Anne K. Voss,a,h René P. Zahedi,i James J. Pitt,j Roman Chrast,c Albert Sickmann,i,k Michael T. Ryan,l Gordon K. Smyth,e,f,h b2 a,h,m,n3 David R. Thorburn, and Hamish S. Scott Downloaded from From the aMolecular Medicine Division, gImmunology Division, and eBioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia, the bMurdoch Childrens Research Institute, Royal Children’s Hospital and Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia, the cDépartement de Génétique Médicale, Université de Lausanne, 1005 Lausanne, Switzerland, the dCentre for Reproduction and Development, Monash Institute of Medical Research, Clayton, Victoria 3168, Australia, the hDepartment of Medical Biology
    [Show full text]
  • Mouse Rbm7 Conditional Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Rbm7 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Rbm7 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Rbm7 gene (NCBI Reference Sequence: NM_144948 ; Ensembl: ENSMUSG00000042396 ) is located on Mouse chromosome 9. 5 exons are identified, with the ATG start codon in exon 1 and the TAA stop codon in exon 5 (Transcript: ENSMUST00000170000). Exon 4 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Rbm7 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP23-147I23 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 4 starts from about 43.77% of the coding region. The knockout of Exon 4 will result in frameshift of the gene. The size of intron 3 for 5'-loxP site insertion: 2468 bp, and the size of intron 4 for 3'-loxP site insertion: 859 bp. The size of effective cKO region: ~627 bp. The cKO region does not have any other known gene. Page 1 of 7 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele gRNA region 5' gRNA region 3' 1 4 5 Targeting vector Targeted allele Constitutive KO allele (After Cre recombination) Legends Exon of mouse Rbm7 Homology arm cKO region loxP site Page 2 of 7 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • Biallelic Alteration and Dysregulation of the Hippo Pathway in Mucinous Tubular and Spindle Cell Carcinoma of the Kidney
    Published OnlineFirst September 7, 2016; DOI: 10.1158/2159-8290.CD-16-0267 RESEARCH BRIEF Biallelic Alteration and Dysregulation of the Hippo Pathway in Mucinous Tubular and Spindle Cell Carcinoma of the Kidney Rohit Mehra 1 , 2 , 3 , Pankaj Vats 1 , 3 , 4 , Marcin Cieslik 3 , Xuhong Cao 3 , 5 , Fengyun Su 3 , Sudhanshu Shukla 3 , Aaron M. Udager 1 , Rui Wang 3 , Jincheng Pan 6 , Katayoon Kasaian 3 , Robert Lonigro 3 , Javed Siddiqui 3 , Kumpati Premkumar 4 , Ganesh Palapattu 7 , Alon Weizer 2 , 7 , Khaled S. Hafez 7 , J. Stuart Wolf Jr 7 , Ankur R. Sangoi 8 , Kiril Trpkov 9 , Adeboye O. Osunkoya 10 , Ming Zhou 11 , Giovanna A. Giannico 12 , Jesse K. McKenney 13 , Saravana M. Dhanasekaran 1 , 3 , and Arul M. Chinnaiyan 1 , 2 , 3 , 5 , 7 ABSTRACT Mucinous tubular and spindle cell carcinoma (MTSCC) is a relatively rare subtype of renal cell carcinoma (RCC) with distinctive morphologic and cytogenetic fea- tures. Here, we carry out whole-exome and transcriptome sequencing of a multi-institutional cohort of MTSCC (n = 22). We demonstrate the presence of either biallelic loss of Hippo pathway tumor sup- pressor genes (TSG) and/or evidence of alteration of Hippo pathway genes in 85% of samples. PTPN14 (31%) and NF2 (22%) were the most commonly implicated Hippo pathway genes, whereas other genes such as SAV1 and HIPK2 were also involved in a mutually exclusive fashion. Mutations in the context of recurrent chromosomal losses amounted to biallelic alterations in these TSGs. As a readout of Hippo pathway inactivation, a majority of cases (90%) exhibited increased nuclear YAP1 protein expression.
    [Show full text]
  • Structural Basis for MTR4–ZCCHC8 Interactions That Stimulate the MTR4 Helicase in the Nuclear Exosome-Targeting Complex
    Structural basis for MTR4–ZCCHC8 interactions that stimulate the MTR4 helicase in the nuclear exosome-targeting complex M. Rhyan Punoa,b and Christopher D. Limaa,b,1 aStructural Biology Program, Sloan Kettering Institute, New York, NY 10065; and bHoward Hughes Medical Institute, Sloan Kettering Institute, New York, NY 10065 Edited by Lynne E. Maquat, University of Rochester School of Medicine and Dentistry, Rochester, NY, and approved May 9, 2018 (received for review February 27, 2018) The nuclear exosome-targeting (NEXT) complex functions as an complex (PPC) (17). NEXT is a nucleoplasmic ternary complex RNA exosome cofactor and is involved in surveillance and turnover consisting of RBM7, ZCCHC8, and MTR4 (15). RBM7 is a of aberrant transcripts and noncoding RNAs. NEXT is a ternary 31-kDa protein with a polyuridine-specific RNA recognition motif complex composed of the RNA-binding protein RBM7, the scaffold (RRM) (18), while the zinc-knuckle protein ZCCHC8 acts as a zinc-knuckle protein ZCCHC8, and the helicase MTR4. While RNA scaffold, mediating the interaction between RBM7 and MTR4 (16). interactions with RBM7 are known, it remains unclear how NEXT NEXT is involved in exosome-mediated surveillance and decay of subunits collaborate to recognize and prepare substrates for degra- noncoding RNAs, such as enhancer RNAs (eRNAs) and aberrant dation. Here, we show that MTR4 helicase activity is enhanced when 3′-extended transcripts from small nuclear RNA (snRNA), telo- associated with RBM7 and ZCCHC8. While uridine-rich substrates merase RNA, and replication-dependent histone genes (15, 19, 20). interact with RBM7 and are preferred, optimal activity is observed It ensures a unidirectional output from pervasive transcription of ′ when substrates include a polyadenylated 3 end.
    [Show full text]
  • Loss of the RNA Helicase SKIV2L2 Impairs Mitotic Progression and Replication-Dependent Histone Mrna Turnover in Murine Cell Lines
    Downloaded from rnajournal.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press Loss of the RNA helicase SKIV2L2 impairs mitotic progression and replication-dependent histone mRNA turnover in murine cell lines ALEXIS M. ONDERAK and JAMES T. ANDERSON Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin 53201, USA ABSTRACT RNA surveillance via the nuclear exosome requires cofactors such as the helicase SKIV2L2 to process and degrade certain noncoding RNAs. This research aimed to characterize the phenotype associated with RNAi knockdown of Skiv2l2 in two murine cancer cell lines: Neuro2A and P19. SKIV2L2 depletion in Neuro2A and P19 cells induced changes in gene expression indicative of cell differentiation and reduced cellular proliferation by 30%. Propidium iodide-based cell-cycle analysis of Skiv2l2 knockdown cells revealed defective progression through the G2/M phase and an accumulation of mitotic cells, suggesting SKIV2L2 contributes to mitotic progression. Since SKIV2L2 targets RNAs to the nuclear exosome for processing and degradation, we identified RNA targets elevated in cells depleted of SKIV2L2 that could account for the observed twofold increase in mitotic cells. Skiv2l2 knockdown cells accumulated replication-dependent histone mRNAs, among other RNAs, that could impede mitotic progression and indirectly trigger differentiation. Keywords: histones; RNA turnover; RNA surveillance; TRAMP complex INTRODUCTION bound by Air1/2p (Schmidt and Butler 2013). The resulting A tail not only distinguishes exosome substrates from RNAs During the course of transcription, nuclear RNA surveillance stabilized by a longer poly(A) tail (Andersen et al. 2008), but orchestrates the proper processing and turnover of certain ′ also generates the 3 overhang necessary for the helicase RNAs.
    [Show full text]
  • Improved Detection of Gene Fusions by Applying Statistical Methods Reveals New Oncogenic RNA Cancer Drivers
    bioRxiv preprint doi: https://doi.org/10.1101/659078; this version posted June 3, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Improved detection of gene fusions by applying statistical methods reveals new oncogenic RNA cancer drivers Roozbeh Dehghannasiri1, Donald Eric Freeman1,2, Milos Jordanski3, Gillian L. Hsieh1, Ana Damljanovic4, Erik Lehnert4, Julia Salzman1,2,5* Author affiliation 1Department of Biochemistry, Stanford University, Stanford, CA 94305 2Department of Biomedical Data Science, Stanford University, Stanford, CA 94305 3Department of Computer Science, University of Belgrade, Belgrade, Serbia 4Seven Bridges Genomics, Cambridge, MA 02142 5Stanford Cancer Institute, Stanford, CA 94305 *Corresponding author [email protected] Short Abstract: The extent to which gene fusions function as drivers of cancer remains a critical open question. Current algorithms do not sufficiently identify false-positive fusions arising during library preparation, sequencing, and alignment. Here, we introduce a new algorithm, DEEPEST, that uses statistical modeling to minimize false-positives while increasing the sensitivity of fusion detection. In 9,946 tumor RNA-sequencing datasets from The Cancer Genome Atlas (TCGA) across 33 tumor types, DEEPEST identifies 31,007 fusions, 30% more than identified by other methods, while calling ten-fold fewer false-positive fusions in non-transformed human tissues. We leverage the increased precision of DEEPEST to discover new cancer biology. For example, 888 new candidate oncogenes are identified based on over-representation in DEEPEST-Fusion calls, and 1,078 previously unreported fusions involving long intergenic noncoding RNAs partners, demonstrating a previously unappreciated prevalence and potential for function.
    [Show full text]
  • RBM7 Subunit of the NEXT Complex Binds U-Rich Sequences and Targets
    4236–4248 Nucleic Acids Research, 2015, Vol. 43, No. 8 Published online 6 April 2015 doi: 10.1093/nar/gkv240 RBM7 subunit of the NEXT complex binds U-rich sequences and targets 3-end extended forms of snRNAs Dominika Hrossova1,2, Tomas Sikorsky1,2, David Potesil1, Marek Bartosovic1,2, Josef Pasulka1, Zbynek Zdrahal1, Richard Stefl1,2,* and Stepanka Vanacova1,* 1CEITEC–Central European Institute of Technology, Masaryk University, Brno, 62500, Czech Republic and 2National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic Downloaded from https://academic.oup.com/nar/article/43/8/4236/2414277 by guest on 01 October 2021 Received October 15, 2014; Revised March 05, 2015; Accepted March 06, 2015 ABSTRACT budding yeast are the Trf4/5-Air1/2-Mtr4 Polyadenylating (TRAMP) and Nrd1-Nab3-Sen1 (NNS) complexes (3–6). The Nuclear Exosome Targeting (NEXT) complex The TRAMP is composed of the non-canonical poly(A)- is a key cofactor of the mammalian nuclear exo- polymerase (PAP) Trf4p or Trf5p, the zinc-knuckle (ZnK) some in the removal of Promoter Upstream Tran- protein Air1p or Air2p and the RNA helicase Mtr4p (4– scripts (PROMPTs) and potentially aberrant forms of 7). TRAMP adds untemplated oligo(A) tail to the 3 end of other noncoding RNAs, such as snRNAs. NEXT is the RNA substrate which serves as a landing platform for composed of three subunits SKIV2L2, ZCCHC8 and the exosome. The NNS is formed by two RNA-binding pro- RBM7. We have recently identified the NEXT complex teins Nrd1p and Nab3p and an RNA helicase Sen1p (8).
    [Show full text]
  • Nº Ref Uniprot Proteína Péptidos Identificados Por MS/MS 1 P01024
    Document downloaded from http://www.elsevier.es, day 26/09/2021. This copy is for personal use. Any transmission of this document by any media or format is strictly prohibited. Nº Ref Uniprot Proteína Péptidos identificados 1 P01024 CO3_HUMAN Complement C3 OS=Homo sapiens GN=C3 PE=1 SV=2 por 162MS/MS 2 P02751 FINC_HUMAN Fibronectin OS=Homo sapiens GN=FN1 PE=1 SV=4 131 3 P01023 A2MG_HUMAN Alpha-2-macroglobulin OS=Homo sapiens GN=A2M PE=1 SV=3 128 4 P0C0L4 CO4A_HUMAN Complement C4-A OS=Homo sapiens GN=C4A PE=1 SV=1 95 5 P04275 VWF_HUMAN von Willebrand factor OS=Homo sapiens GN=VWF PE=1 SV=4 81 6 P02675 FIBB_HUMAN Fibrinogen beta chain OS=Homo sapiens GN=FGB PE=1 SV=2 78 7 P01031 CO5_HUMAN Complement C5 OS=Homo sapiens GN=C5 PE=1 SV=4 66 8 P02768 ALBU_HUMAN Serum albumin OS=Homo sapiens GN=ALB PE=1 SV=2 66 9 P00450 CERU_HUMAN Ceruloplasmin OS=Homo sapiens GN=CP PE=1 SV=1 64 10 P02671 FIBA_HUMAN Fibrinogen alpha chain OS=Homo sapiens GN=FGA PE=1 SV=2 58 11 P08603 CFAH_HUMAN Complement factor H OS=Homo sapiens GN=CFH PE=1 SV=4 56 12 P02787 TRFE_HUMAN Serotransferrin OS=Homo sapiens GN=TF PE=1 SV=3 54 13 P00747 PLMN_HUMAN Plasminogen OS=Homo sapiens GN=PLG PE=1 SV=2 48 14 P02679 FIBG_HUMAN Fibrinogen gamma chain OS=Homo sapiens GN=FGG PE=1 SV=3 47 15 P01871 IGHM_HUMAN Ig mu chain C region OS=Homo sapiens GN=IGHM PE=1 SV=3 41 16 P04003 C4BPA_HUMAN C4b-binding protein alpha chain OS=Homo sapiens GN=C4BPA PE=1 SV=2 37 17 Q9Y6R7 FCGBP_HUMAN IgGFc-binding protein OS=Homo sapiens GN=FCGBP PE=1 SV=3 30 18 O43866 CD5L_HUMAN CD5 antigen-like OS=Homo
    [Show full text]
  • Caracterización De Complejos CDK-Ciclina Atípicos Humanos Eva Quandt Herrera
    Caracterización de complejos CDK-Ciclina atípicos humanos Eva Quandt Herrera ADVERTIMENT. La consulta d’aquesta tesi queda condicionada a l’acceptació de les següents condicions d'ús: La difusió d’aquesta tesi per mitjà del servei TDX (www.tesisenxarxa.net) ha estat autoritzada pels titulars dels drets de propietat intel·lectual únicament per a usos privats emmarcats en activitats d’investigació i docència. No s’autoritza la seva reproducció amb finalitats de lucre ni la seva difusió i posada a disposició des d’un lloc aliè al servei TDX. No s’autoritza la presentació del seu contingut en una finestra o marc aliè a TDX (framing). Aquesta reserva de drets afecta tant al resum de presentació de la tesi com als seus continguts. En la utilització o cita de parts de la tesi és obligat indicar el nom de la persona autora. ADVERTENCIA. La consulta de esta tesis queda condicionada a la aceptación de las siguientes condiciones de uso: La difusión de esta tesis por medio del servicio TDR (www.tesisenred.net) ha sido autorizada por los titulares de los derechos de propiedad intelectual únicamente para usos privados enmarcados en actividades de investigación y docencia. No se autoriza su reproducción con finalidades de lucro ni su difusión y puesta a disposición desde un sitio ajeno al servicio TDR. No se autoriza la presentación de su contenido en una ventana o marco ajeno a TDR (framing). Esta reserva de derechos afecta tanto al resumen de presentación de la tesis como a sus contenidos. En la utilización o cita de partes de la tesis es obligado indicar el nombre de la persona autora.
    [Show full text]
  • A High Throughput, Functional Screen of Human Body Mass Index GWAS Loci Using Tissue-Specific Rnai Drosophila Melanogaster Crosses Thomas J
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2018 A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses Thomas J. Baranski Washington University School of Medicine in St. Louis Aldi T. Kraja Washington University School of Medicine in St. Louis Jill L. Fink Washington University School of Medicine in St. Louis Mary Feitosa Washington University School of Medicine in St. Louis Petra A. Lenzini Washington University School of Medicine in St. Louis See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Baranski, Thomas J.; Kraja, Aldi T.; Fink, Jill L.; Feitosa, Mary; Lenzini, Petra A.; Borecki, Ingrid B.; Liu, Ching-Ti; Cupples, L. Adrienne; North, Kari E.; and Province, Michael A., ,"A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses." PLoS Genetics.14,4. e1007222. (2018). https://digitalcommons.wustl.edu/open_access_pubs/6820 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]. Authors Thomas J. Baranski, Aldi T. Kraja, Jill L. Fink, Mary Feitosa, Petra A. Lenzini, Ingrid B. Borecki, Ching-Ti Liu, L. Adrienne Cupples, Kari E. North, and Michael A. Province This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/6820 RESEARCH ARTICLE A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses Thomas J.
    [Show full text]