A Novel Role for the Chromatin Remodeling Atpase Brg1 During Zygotic Genome Activation

Total Page:16

File Type:pdf, Size:1020Kb

A Novel Role for the Chromatin Remodeling Atpase Brg1 During Zygotic Genome Activation A novel role for the chromatin remodeling ATPase Brg1 during zygotic genome activation in Xenopus Dissertation Zur Erlangung des akademischen Grades Doctor rerum naturalis (Dr. rer.nat.) Vorgelegt der Fakultät für Biologie der Ludwigs-Maximilians-Universität zu München Gabriele Maria Wagner aus Heidelberg München 2014 Tag der Einreichung: 20. Mai 2014 Tag der mündlichen Prüfung: 31. Juli 2014 Erstgutachter: Prof. Dr. Peter Becker Zweitgutachter: Prof. Dr. Hans Straka Drittgutachter: Prof. Dr. Angelika Böttger Viertgutachter: Prof. Dr. John Parsch Eidesstattliche Erklärung Ich versichere hiermit an Eides statt, dass die vorgelegte Dissertation von mir selbständig und ohne unerlaubte Hilfe angefertigt ist. München, den …………………. ............................................................ -Unterschrift- Table of contents Zusammenfassung ............................................................................................................. I Summary ......................................................................................................................... III 1 Introduction ........................................................................................................... 1 1.1 Xenopus as a model organism ................................................................................................. 1 1.1.1 Life cycle of Xenopus ........................................................................................................... 1 1.1.2 The zygotic genome activation ............................................................................................ 2 1.1.3 Axial patterning during Xenopus development ................................................................... 4 1.2 Chromatin dynamics ................................................................................................................ 8 1.2.1 Chromatin structure ............................................................................................................ 8 1.2.2 Posttranslational modifications ......................................................................................... 10 1.2.3 Chromatin remodeling complexes .................................................................................... 12 1.3 The SWI/SNF complex in development ................................................................................. 13 1.4 Objectives .............................................................................................................................. 15 2 Materials and Methods ........................................................................................ 17 2.1 Materials ................................................................................................................................ 17 2.1.1 Animals .............................................................................................................................. 17 2.1.2 Bacteria .............................................................................................................................. 17 2.1.3 Enzymes ............................................................................................................................. 17 2.1.4 Chemicals ........................................................................................................................... 18 2.1.5 Antibodies.......................................................................................................................... 18 2.1.6 Oligonucleotides ................................................................................................................ 19 2.1.7 Plasmids ............................................................................................................................. 23 2.1.8 Equipment ......................................................................................................................... 24 2.1.9 Software ............................................................................................................................ 25 2.2 Methods ................................................................................................................................ 25 2.2.1 DNA standard methods ..................................................................................................... 25 2.2.2 RNA standard methods ..................................................................................................... 29 2.2.3 Protein analysis ................................................................................................................. 34 2.2.4 Embryological methods ..................................................................................................... 36 2.2.5 Embryological explantations ............................................................................................. 39 3 Results ................................................................................................................. 41 3.1 Comparison of morpholino oligonucleotides ........................................................................ 41 3.2 BMO1 reduces Brg1 protein level from MBT on ................................................................... 42 3.3 Morphological gain- and loss-of-function studies ................................................................. 44 3.3.1 Targeted loss-of-function analysis..................................................................................... 44 3.3.2 Targeted gain-of-function analysis .................................................................................... 45 3.4 Wnt signaling is not affected in Brg1 morphants .................................................................. 46 3.5 Axis patterning genes are downregulated in neurula stage ................................................. 48 3.6 Dorso-ventral patterning in gastrula embryos is perturbed ................................................. 50 3.7 Brg1 knockdown affects gene expression already at blastula .............................................. 52 3.8 ß-catenin target gene siamois is not changed upon loss of Brg1.......................................... 54 3.9 Exogenous chordin mRNA can compensate for the loss of Brg1 .......................................... 55 3.10 A Brg1 morphant BCNE causes loss of eye and brain structures .......................................... 57 3.11 Molecular analysis of BCNE transplants ................................................................................ 58 3.12 Brg1 knockdown impairs the transcriptional readout of wnt-and nodal inducers ............... 59 3.13 Genes outside the BCNE are affected by Brg1 knockdown................................................... 61 3.14 Brg1 knockdown in Xenopus tropicalis .................................................................................. 63 3.15 BMO1 and BMO2 reduce Brg1 protein level at blastula and cause embryonic lethality during gastrulation ................................................................................................................................ 64 3.16 Morphant embryos are not delayed in development and do not activate apoptosis .......... 66 3.17 Genome-wide transcriptome analysis of BMO1 morphant embryos ................................... 67 3.17.1 Genes related to “nervous system development” are affected in Brg1 morphants ........ 68 3.17.2 BMO1 affects a broad range of genes throughout all germ layers ................................... 69 3.18 Brg1 knockdown by BMO2 recapitulates the major changes in gene expression ................ 71 3.19 Global transcription changes at midblastula transition ........................................................ 73 3.20 Comparison of pre- and postMBT transcriptomes ................................................................ 74 3.21 Highly upregulated genes at MBT need Brg1 protein ........................................................... 75 4 Discussion ............................................................................................................ 79 4.1 Morpholino efficiency and targeting behavior ..................................................................... 80 4.2 Phenotypic analysis of Brg1 morphant embryos .................................................................. 81 4.2.1 Morpholino based knockdown analyses ........................................................................... 81 4.2.2 Overexpression of wild type Brg1 ..................................................................................... 83 4.3 Genome-wide transcriptome analysis of Brg1 morphant embryos ...................................... 84 4.4 Brg1 and its role in axes determination ................................................................................ 86 4.4.1 Antero-posterior axis ......................................................................................................... 86 4.4.2 Dorso-ventral patterning ................................................................................................... 87 4.5 Brg1 and its role in neurogenesis .......................................................................................... 90 4.6 Brg1 and its role during zygotic genome activation .............................................................. 92 4.7 Possible Brg1 mechanism at the burst of transcription ........................................................ 94 4.8 Conclusions and future directions ........................................................................................
Recommended publications
  • An Order Estimation Based Approach to Identify Response Genes
    AN ORDER ESTIMATION BASED APPROACH TO IDENTIFY RESPONSE GENES FOR MICRO ARRAY TIME COURSE DATA A Thesis Presented to The Faculty of Graduate Studies of The University of Guelph by ZHIHENG LU In partial fulfilment of requirements for the degree of Doctor of Philosophy September, 2008 © Zhiheng Lu, 2008 Library and Bibliotheque et 1*1 Archives Canada Archives Canada Published Heritage Direction du Branch Patrimoine de I'edition 395 Wellington Street 395, rue Wellington Ottawa ON K1A0N4 Ottawa ON K1A0N4 Canada Canada Your file Votre reference ISBN: 978-0-494-47605-5 Our file Notre reference ISBN: 978-0-494-47605-5 NOTICE: AVIS: The author has granted a non­ L'auteur a accorde une licence non exclusive exclusive license allowing Library permettant a la Bibliotheque et Archives and Archives Canada to reproduce, Canada de reproduire, publier, archiver, publish, archive, preserve, conserve, sauvegarder, conserver, transmettre au public communicate to the public by par telecommunication ou par Plntemet, prefer, telecommunication or on the Internet, distribuer et vendre des theses partout dans loan, distribute and sell theses le monde, a des fins commerciales ou autres, worldwide, for commercial or non­ sur support microforme, papier, electronique commercial purposes, in microform, et/ou autres formats. paper, electronic and/or any other formats. The author retains copyright L'auteur conserve la propriete du droit d'auteur ownership and moral rights in et des droits moraux qui protege cette these. this thesis. Neither the thesis Ni la these ni des extraits substantiels de nor substantial extracts from it celle-ci ne doivent etre imprimes ou autrement may be printed or otherwise reproduits sans son autorisation.
    [Show full text]
  • Further Delineation of Chromosomal Consensus Regions in Primary
    Leukemia (2007) 21, 2463–2469 & 2007 Nature Publishing Group All rights reserved 0887-6924/07 $30.00 www.nature.com/leu ORIGINAL ARTICLE Further delineation of chromosomal consensus regions in primary mediastinal B-cell lymphomas: an analysis of 37 tumor samples using high-resolution genomic profiling (array-CGH) S Wessendorf1,6, TFE Barth2,6, A Viardot1, A Mueller3, HA Kestler3, H Kohlhammer1, P Lichter4, M Bentz5,HDo¨hner1,PMo¨ller2 and C Schwaenen1 1Klinik fu¨r Innere Medizin III, Zentrum fu¨r Innere Medizin der Universita¨t Ulm, Ulm, Germany; 2Institut fu¨r Pathologie, Universita¨t Ulm, Ulm, Germany; 3Forschungsdozentur Bioinformatik, Universita¨t Ulm, Ulm, Germany; 4Abt. Molekulare Genetik, Deutsches Krebsforschungszentrum, Heidelberg, Germany and 5Sta¨dtisches Klinikum Karlsruhe, Karlsruhe, Germany Primary mediastinal B-cell lymphoma (PMBL) is an aggressive the expression of BSAP, BOB1, OCT2, PAX5 and PU1 was extranodal B-cell non-Hodgkin’s lymphoma with specific clin- added to the spectrum typical of PMBL features.9 ical, histopathological and genomic features. To characterize Genetically, a pattern of highly recurrent karyotype alterations further the genotype of PMBL, we analyzed 37 tumor samples and PMBL cell lines Med-B1 and Karpas1106P using array- with the hallmark of chromosomal gains of the subtelomeric based comparative genomic hybridization (matrix- or array- region of chromosome 9 supported the concept of a unique CGH) to a 2.8k genomic microarray. Due to a higher genomic disease entity that distinguishes PMBL from other B-cell non- resolution, we identified altered chromosomal regions in much Hodgkin’s lymphomas.10,11 Together with less specific gains on higher frequencies compared with standard CGH: for example, 2p15 and frequent mutations of the SOCS1 gene, a notable þ 9p24 (68%), þ 2p15 (51%), þ 7q22 (32%), þ 9q34 (32%), genomic similarity to classical Hodgkin’s lymphoma was þ 11q23 (18%), þ 12q (30%) and þ 18q21 (24%).
    [Show full text]
  • Human Transcription Factor Protein-Protein Interactions in Health and Disease
    HELKA GÖÖS GÖÖS HELKA Recent Publications in this Series 45/2019 Mgbeahuruike Eunice Ego Evaluation of the Medicinal Uses and Antimicrobial Activity of Piper guineense (Schumach & Thonn) 46/2019 Suvi Koskinen AND DISEASE IN HEALTH INTERACTIONS PROTEIN-PROTEIN FACTOR HUMAN TRANSCRIPTION Near-Occlusive Atherosclerotic Carotid Artery Disease: Study with Computed Tomography Angiography 47/2019 Flavia Fontana DISSERTATIONES SCHOLAE DOCTORALIS AD SANITATEM INVESTIGANDAM Biohybrid Cloaked Nanovaccines for Cancer Immunotherapy UNIVERSITATIS HELSINKIENSIS 48/2019 Marie Mennesson Kainate Receptor Auxiliary Subunits Neto1 and Neto2 in Anxiety and Fear-Related Behaviors 49/2019 Zehua Liu Porous Silicon-Based On-Demand Nanohybrids for Biomedical Applications 50/2019 Veer Singh Marwah Strategies to Improve Standardization and Robustness of Toxicogenomics Data Analysis HELKA GÖÖS 51/2019 Iryna Hlushchenko Actin Regulation in Dendritic Spines: From Synaptic Plasticity to Animal Behavior and Human HUMAN TRANSCRIPTION FACTOR PROTEIN-PROTEIN Neurodevelopmental Disorders 52/2019 Heini Liimatta INTERACTIONS IN HEALTH AND DISEASE Efectiveness of Preventive Home Visits among Community-Dwelling Older People 53/2019 Helena Karppinen Older People´s Views Related to Their End of Life: Will-to-Live, Wellbeing and Functioning 54/2019 Jenni Laitila Elucidating Nebulin Expression and Function in Health and Disease 55/2019 Katarzyna Ciuba Regulation of Contractile Actin Structures in Non-Muscle Cells 56/2019 Sami Blom Spatial Characterisation of Prostate Cancer by Multiplex
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Sequence Analysis of Familial Neurodevelopmental Disorders
    SEQUENCE ANALYSIS OF FAMILIAL NEURODEVELOPMENTAL DISORDERS by Joseph Mark Tilghman A dissertation submitted to Johns Hopkins University in conformity with the requirements for the degree of Doctor of Philosophy Baltimore, Maryland December 2020 © 2020 Joseph Tilghman All Rights Reserved Abstract: In the practice of human genetics, there is a gulf between the study of Mendelian and complex inheritance. When diagnosis of families affected by presumed monogenic syndromes is undertaken by genomic sequencing, these families are typically considered to have been solved only when a single gene or variant showing apparently Mendelian inheritance is discovered. However, about half of such families remain unexplained through this approach. On the other hand, common regulatory variants conferring low risk of disease still predominate our understanding of individual disease risk in complex disorders, despite rapidly increasing access to rare variant genotypes through sequencing. This dissertation utilizes primarily exome sequencing across several developmental disorders (having different levels of genetic complexity) to investigate how to best use an individual’s combination of rare and common variants to explain genetic risk, phenotypic heterogeneity, and the molecular bases of disorders ranging from those presumed to be monogenic to those known to be highly complex. The study described in Chapter 2 addresses putatively monogenic syndromes, where we used exome sequencing of four probands having syndromic neurodevelopmental disorders from an Israeli-Arab founder population to diagnose recessive and dominant disorders, highlighting the need to consider diverse modes of inheritance and phenotypic heterogeneity. In the study described in Chapter 3, we address the case of a relatively tractable multifactorial disorder, Hirschsprung disease.
    [Show full text]
  • UNIVERSITY of CALIFORNIA, IRVINE Combinatorial Regulation By
    UNIVERSITY OF CALIFORNIA, IRVINE Combinatorial regulation by maternal transcription factors during activation of the endoderm gene regulatory network DISSERTATION submitted in partial satisfaction of the requirements for the degree of DOCTOR OF PHILOSOPHY in Biological Sciences by Kitt D. Paraiso Dissertation Committee: Professor Ken W.Y. Cho, Chair Associate Professor Olivier Cinquin Professor Thomas Schilling 2018 Chapter 4 © 2017 Elsevier Ltd. © 2018 Kitt D. Paraiso DEDICATION To the incredibly intelligent and talented people, who in one way or another, helped complete this thesis. ii TABLE OF CONTENTS Page LIST OF FIGURES vii LIST OF TABLES ix LIST OF ABBREVIATIONS X ACKNOWLEDGEMENTS xi CURRICULUM VITAE xii ABSTRACT OF THE DISSERTATION xiv CHAPTER 1: Maternal transcription factors during early endoderm formation in 1 Xenopus Transcription factors co-regulate in a cell type-specific manner 2 Otx1 is expressed in a variety of cell lineages 4 Maternal otx1 in the endodermal conteXt 5 Establishment of enhancers by maternal transcription factors 9 Uncovering the endodermal gene regulatory network 12 Zygotic genome activation and temporal control of gene eXpression 14 The role of maternal transcription factors in early development 18 References 19 CHAPTER 2: Assembly of maternal transcription factors initiates the emergence 26 of tissue-specific zygotic cis-regulatory regions Introduction 28 Identification of maternal vegetally-localized transcription factors 31 Vegt and OtX1 combinatorially regulate the endodermal 33 transcriptome iii
    [Show full text]
  • Viewed in [2, 3])
    Yildiz et al. Neural Development (2019) 14:5 https://doi.org/10.1186/s13064-019-0129-x RESEARCH ARTICLE Open Access Zebrafish prdm12b acts independently of nkx6.1 repression to promote eng1b expression in the neural tube p1 domain Ozge Yildiz1, Gerald B. Downes2 and Charles G. Sagerström1* Abstract Background: Functioning of the adult nervous system depends on the establishment of neural circuits during embryogenesis. In vertebrates, neurons that make up motor circuits form in distinct domains along the dorsoventral axis of the neural tube. Each domain is characterized by a unique combination of transcription factors (TFs) that promote a specific fate, while repressing fates of adjacent domains. The prdm12 TF is required for the expression of eng1b and the generation of V1 interneurons in the p1 domain, but the details of its function remain unclear. Methods: We used CRISPR/Cas9 to generate the first germline mutants for prdm12 and employed this resource, together with classical luciferase reporter assays and co-immunoprecipitation experiments, to study prdm12b function in zebrafish. We also generated germline mutants for bhlhe22 and nkx6.1 to examine how these TFs act with prdm12b to control p1 formation. Results: We find that prdm12b mutants lack eng1b expression in the p1 domain and also possess an abnormal touch-evoked escape response. Using luciferase reporter assays, we demonstrate that Prdm12b acts as a transcriptional repressor. We also show that the Bhlhe22 TF binds via the Prdm12b zinc finger domain to form a complex. However, bhlhe22 mutants display normal eng1b expression in the p1 domain. While prdm12 has been proposed to promote p1 fates by repressing expression of the nkx6.1 TF, we do not observe an expansion of the nkx6.1 domain upon loss of prdm12b function, nor is eng1b expression restored upon simultaneous loss of prdm12b and nkx6.1.
    [Show full text]
  • Sexual Dimorphism in the Meiotic Requirement for PRDM9: a Mammalian Evolutionary
    bioRxiv preprint doi: https://doi.org/10.1101/2020.03.10.985358; this version posted March 13, 2020. 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. 1 1 Sexual dimorphism in the meiotic requirement for PRDM9: a mammalian evolutionary 2 safeguard 3 Short title: 4 Sex-limited requirement of PRDM9 in mice 5 One Sentence Summary: 6 Sex-specific modulation of a meiotic DNA damage checkpoint limits the requirement for PRDM9 in 7 mammalian fertility. 8 9 Authors 10 Natalie R Powers1, Beth L Dumont1, Chihiro Emori1, Raman Akinyanju Lawal1, Catherine Brunton1, Ken 11 Paigen1, Mary Ann Handel1, Ewelina Bolcun-Filas1, Petko M Petkov1, and Tanmoy Bhattacharyya1,* 12 1. The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA. 13 *Correspondence to: [email protected] 14 Abstract: 15 In many mammals, genomic sites for recombination are determined by histone methyltransferase PRMD9. Mice 16 lacking PRDM9 are infertile, but instances of fertility or semi-fertility in the absence of PRDM9 have been 17 reported in mice, canines and a human female. Such findings raise the question of how the loss of PRDM9 is 18 circumvented to maintain reproductive fitness. We show that genetic background and sex-specific modifiers can 19 obviate the requirement for PRDM9 in mice. Specifically, the meiotic DNA damage checkpoint protein CHK2 20 acts as a modifier allowing female-specific fertility in the absence of PRDM9. We also report that in the 21 absence of PRDM9, a PRDM9-independent recombination system is compatible with female meiosis and 22 fertility, suggesting sex-specific regulation of meiotic recombination, a finding with implications for speciation.
    [Show full text]
  • DULIP: a Dual Luminescence-Based Co-Immunoprecipitation Assay for Interactome Mapping in Mammalian Cells
    Repository of the Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association http://edoc.mdc-berlin.de/14998 DULIP: A dual luminescence-based co-immunoprecipitation assay for interactome mapping in mammalian cells Trepte, P., Buntru, A., Klockmeier, K., Willmore, L., Arumughan, A., Secker, C., Zenkner, M., Brusendorf, L., Rau, K., Redel, A., Wanker, E.E. NOTICE: this is the author’s version of a work that was accepted for publication in the Journal of Molecular Biology. Changes resulting from the publishing process, such as peer review, editing, corrections, structural formatting, and other quality control mechanisms may not be reflected in this document. Changes may have been made to this work since it was submitted for publication. A definitive version was subsequently published in: Journal of Molecular Biology 2015 MMM DD ; 427(21): 3375-3388 doi: 10.1016/j.jmb.2015.08.003 Publisher: Elsevier © 2015, Elsevier. This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/ or send a letter to Creative Commons, PO Box 1866, Mountain View, CA 94042, USA. DULIP: A DUAL LUMINESCENCE-BASED CO-IMMUNOPRECIPITATION ASSAY FOR INTERACTOME MAPPING IN MAMMALIAN CELLS Philipp Treptea, Alexander Buntrua#, Konrad Klockmeiera#, Lindsay Willmorea, Anup Arumughana, Christopher Seckera, Martina Zenknera, Lydia Brusendorfa, Kirstin Raua, Alexandra Redela and Erich E Wankera* a Neuroproteomics, Max Delbrueck Center for Molecular Medicine, Robert-Roessle- Straße 10, 13125 Berlin, Germany # Contributed equally * Corresponding author, E-mail address: [email protected], Telephone: +49- 30-9406-2157, Fax: +49-30-9406-2552 ABSTRACT Mapping of protein-protein interactions (PPIs) is critical for understanding protein function and complex biological processes.
    [Show full text]
  • Supplementary Figure S4
    18DCIS 18IDC Supplementary FigureS4 22DCIS 22IDC C D B A E (0.77) (0.78) 16DCIS 14DCIS 28DCIS 16IDC 28IDC (0.43) (0.49) 0 ADAMTS12 (p.E1469K) 14IDC ERBB2, LASP1,CDK12( CCNE1 ( NUTM2B SDHC,FCGR2B,PBX1,TPR( CD1D, B4GALT3, BCL9, FLG,NUP21OL,TPM3,TDRD10,RIT1,LMNA,PRCC,NTRK1 0 ADAMTS16 (p.E67K) (0.67) (0.89) (0.54) 0 ARHGEF38 (p.P179Hfs*29) 0 ATG9B (p.P823S) (0.68) (1.0) ARID5B, CCDC6 CCNE1, TSHZ3,CEP89 CREB3L2,TRIM24 BRAF, EGFR (7p11); 0 ABRACL (p.R35H) 0 CATSPER1 (p.P152H) 0 ADAMTS18 (p.Y799C) 19q12 0 CCDC88C (p.X1371_splice) (0) 0 ADRA1A (p.P327L) (10q22.3) 0 CCNF (p.D637N) −4 −2 −4 −2 0 AKAP4 (p.G454A) 0 CDYL (p.Y353Lfs*5) −4 −2 Log2 Ratio Log2 Ratio −4 −2 Log2 Ratio Log2 Ratio 0 2 4 0 2 4 0 ARID2 (p.R1068H) 0 COL27A1 (p.G646E) 0 2 4 0 2 4 2 EDRF1 (p.E521K) 0 ARPP21 (p.P791L) ) 0 DDX11 (p.E78K) 2 GPR101, p.A174V 0 ARPP21 (p.P791T) 0 DMGDH (p.W606C) 5 ANP32B, p.G237S 16IDC (Ploidy:2.01) 16DCIS (Ploidy:2.02) 14IDC (Ploidy:2.01) 14DCIS (Ploidy:2.9) -3 -2 -1 -3 -2 -1 -3 -2 -1 -3 -2 -1 -3 -2 -1 -3 -2 -1 Log Ratio Log Ratio Log Ratio Log Ratio 12DCIS 0 ASPM (p.S222T) Log Ratio Log Ratio 0 FMN2 (p.G941A) 20 1 2 3 2 0 1 2 3 2 ERBB3 (p.D297Y) 2 0 1 2 3 20 1 2 3 0 ATRX (p.L1276I) 20 1 2 3 2 0 1 2 3 0 GALNT18 (p.F92L) 2 MAPK4, p.H147Y 0 GALNTL6 (p.E236K) 5 C11orf1, p.Y53C (10q21.2); 0 ATRX (p.R1401W) PIK3CA, p.H1047R 28IDC (Ploidy:2.0) 28DCIS (Ploidy:2.0) 22IDC (Ploidy:3.7) 22DCIS (Ploidy:4.1) 18IDC (Ploidy:3.9) 18DCIS (Ploidy:2.3) 17q12 0 HCFC1 (p.S2025C) 2 LCMT1 (p.S34A) 0 ATXN7L2 (p.X453_splice) SPEN, p.P677Lfs*13 CBFB 1 2 3 4 5 6 7 8 9 10 11
    [Show full text]