Mechanisms Underlying the Temporal and Selective Induction of Ptf1a Target Genes

Total Page:16

File Type:pdf, Size:1020Kb

Mechanisms Underlying the Temporal and Selective Induction of Ptf1a Target Genes Mechanisms underlying the temporal and selective induction of Ptf1a target genes Doctoral Thesis Dissertation for the award of the degree "Doctor rerum naturalium (Dr. rer. nat.)" in the GGNB program "Genes and Development“ at the Georg August University Göttingen Faculty of Biology submitted by Sven Richts born in Werl, Germany Göttingen, December 2017 Members of the Thesis Committee: Dr. Kristine A. Henningfeld (Supervisor, Reviewer) Department of Developmental Biochemistry, University of Göttingen Prof. Dr. Steven Johnsen (Reviewer) Clinic for General, Visceral and Pediatric Surgery, University of Göttingen Prof. Dr. Thomas Dresbach Department for Anatomy and Embryology University of Göttingen Affidavit Herewith, I declare that I prepared the PhD thesis "Mechanisms underlying the temporal and selective induction of Ptf1a target genes " on my own and with no other sources and aids than quoted. Submission date Göttingen, 31.12.2017 _________________________ Sven Richts Table of contents I Table of contents Table of contents .......................................................................................... I Acknowledgements .................................................................................... VI Abstract ...................................................................................................... VII List of figures ............................................................................................ VIII List of tables ................................................................................................. X Abbreviations ............................................................................................. XII 1. Introduction .............................................................................................. 1 1.1 Neurogenesis in Xenopus laevis ....................................................... 1 1.2 Neural induction ................................................................................. 2 1.3 bHLH transcription factors and their role during neuronal differentiation and determination ............................................................ 4 1.4 Lateral inhibition ................................................................................. 8 1.5 Neuronal subtype specification ....................................................... 10 1.6 Role of phosphorylation in regulating neurogenesis .................... 13 1.7 Epigenetic regulation of neurogenesis ........................................... 14 1.8 Changes in the epigenetic landscape during development .......... 19 1.9 Ptf1a ................................................................................................... 20 1.10 Ptf1a forms a trimeric transcription factor complex .................... 23 1.11 Gene induction and regulation by Ptf1a ....................................... 24 1.12 Aims ................................................................................................. 26 2. Materials and Methods .......................................................................... 27 2.1 Materials ............................................................................................ 27 2.1.1 X. laevis ........................................................................................ 27 2.1.2 Bacteria ........................................................................................ 27 2.1.3 Chemicals ..................................................................................... 27 2.1.4 Antibiotics and Media ................................................................... 27 Table of contents II 2.1.5 Oligonucleotides............................................................................ 28 2.1.5.1 RT-PCR oligonucleotides ........................................................ 28 2.1.5.2 Sequencing oligonucleotides .................................................. 29 2.1.6 Sense RNA constructs .................................................................. 30 2.1.7 Antisense RNA constructs ............................................................ 34 2.2 Methods ............................................................................................. 35 2.2.1 In vitro synthesis of capped sense RNA ........................................ 35 2.2.2 Xenopus methods ......................................................................... 35 2.2.2.1 Priming of X. laevis frogs ........................................................ 35 2.2.2.2 Preparation of the testis .......................................................... 36 2.2.2.3 In vitro fertilization ................................................................... 36 2.2.2.4 Microinjection .......................................................................... 36 2.2.2.5 Preparation of ectodermal explants (animal caps) .................. 36 2.2.2.6 Dexamethasone treatment ...................................................... 37 2.2.2.7 Bimolecular Fluorescent Complementation assay (BiFC) ....... 37 2.2.2.8 In vitro synthesis of antisense RNA ........................................ 37 2.2.2.9 Whole mount in-situ hybridization (WMISH) ........................... 38 2.2.2.9.1 Fixation and X-Gal staining............................................... 39 2.2.2.9.2 Rehydration ...................................................................... 39 2.2.2.9.3 Proteinase K treatment ..................................................... 40 2.2.2.9.4 Acetylation ........................................................................ 40 2.2.2.9.5 Hybridization reaction ....................................................... 40 2.2.2.9.6 Washings .......................................................................... 40 2.2.2.9.7 Antibody reaction .............................................................. 41 2.2.2.9.8 Staining reaction ............................................................... 41 2.2.2.9.9 Background removal and bleaching ................................. 41 2.2.3 Agarose gel electrophoresis .......................................................... 41 2.2.4.1 PCR cloning ............................................................................ 42 Table of contents III 2.2.4.2 Gel purification of PCR and restriction fragments .................. 42 2.2.4.3 DNA restriction digestion ........................................................ 42 2.2.4.4 Ligation................................................................................... 42 2.2.4.5 Chemical transformation of bacterial cells .............................. 43 2.2.4.6 Plasmid DNA preparation ....................................................... 43 2.2.4.7 DNA sequencing .................................................................... 43 2.2.5 ATAC sequencing ........................................................................ 44 2.2.5.1 Transposition reaction ............................................................ 44 2.2.5.2 Library preparation ................................................................. 44 2.2.5.3 Sequencing (by TAL) ............................................................. 45 2.2.5.4 Sequencing alignment and processing (performed by TAL)... 45 2.2.6 Gene expression analysis by semiquantitative RT-PCR or quantitative Nanostring analysis ............................................................ 46 2.2.6.1 Total RNA isolation from ectodermal explants and whole embryos ............................................................................................. 46 2.2.6.2.1 Reverse transcription ....................................................... 47 2.2.6.2.2 Reverse transcription-PCR (RT-PCR) ............................. 47 2.2.6.3 Quantitative Nanostring analysis ............................................ 47 2.2.7 RNA sequencing .......................................................................... 48 2.2.7.1 RNA isolation ......................................................................... 48 2.2.7.2 Sample preparation and sequencing ...................................... 49 2.2.7.3 Sequencing alignment (performed by TAL) ............................ 49 2.2.7.4 Statistical analysis (performed by TAL) .................................. 49 3. Results .................................................................................................... 50 3.1 Mutation of a single threonine residue in the Ptf1a C2 domain is sufficient to induce a mixed induced neuronal transmitter phenotype ................................................................................................................. 50 3.2 Introducing a negative charge at the Ptf1aT243 residue induces a more wildtype Ptf1a like phenotype ...................................................... 56 Table of contents IV 3.3 The mixed neuronal transmitter phenotype induced by Ptf1aT243A is not the result of an impaired interaction with Rbpj .......................... 57 3.4 Ptf1aT243 mutants strongly bind to Prdm13 .................................... 58 3.5 Ptf1a induces direct target genes at different time points............. 62 3.6 Temporal expression of Ptf1a target genes by RNA sequencing . 65 3.7 Ptf1a target genes are highly enriched for playing roles during transcription and neurogenesis ............................................................. 67 3.8 A knock-down of Brg1 affects the induction of indirect Ptf1a target genes ........................................................................................................ 69 3.9
Recommended publications
  • TOX2 Is Differentially Expressed in Non-Small Cell Lung Cancer-PDF 092220
    1 Tox2 is differentially expressed in non-small cell lung cancer and associates with patient 2 survival. 3 Shahan Mamoor1 4 [email protected] East Islip, NY USA 5 6 Non-small cell lung cancer (NSCLC) is the leading cause of cancer death in the United States1. 7 We mined published microarray data2,3,4 to identify differentially expressed genes in NSCLC. 8 We found that the gene encoding the thymyocyte selection-associated high mobility box protein 9 family member 2 transcription factor Tox2 was among the genes whose expression was most quantitatively different in tumors from patients with NSCLC as compared to the lung. Tox2 10 expression was significantly decreased in NSCLC tumors as compared to the lung, and lower 11 expression of Tox2 in patient tumors was significantly associated with worse overall survival. Tox2 may be important for initiation or progression of non-small cell lung cancer in humans. 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 Keywords: Tox2, NSCLC, non-small cell lung cancer, systems biology of NSCLC, targeted 27 therapeutics in NSCLC. 28 1 OF 16 1 In 2016, lung cancer resulted in the death of 158,000 Americans; 81% of all patients 2 diagnosed with lung cancer will expire within 5 years5. Non-small cell lung cancer (NSCLC) is 3 4 the most common type of lung cancer, diagnosed in 84% of patients with lung cancer, and 76% 5 of all patients with NSCLC will expire within 5 years5. The rational development of targeted 6 therapeutics to treat patients with NSCLC can be supported by an enhanced understanding of 7 8 fundamental transcriptional features of NSCLC tumors.
    [Show full text]
  • TOX As a Potential Target for Immunotherapy in Lymphocytic Malignancies Chaofeng Liang1,2, Shuxin Huang1, Yujie Zhao1, Shaohua Chen1* and Yangqiu Li1*
    Liang et al. Biomarker Research (2021) 9:20 https://doi.org/10.1186/s40364-021-00275-y REVIEW Open Access TOX as a potential target for immunotherapy in lymphocytic malignancies Chaofeng Liang1,2, Shuxin Huang1, Yujie Zhao1, Shaohua Chen1* and Yangqiu Li1* Abstract TOX (thymocyte selection-associated HMG BOX) is a member of a family of transcriptional factors that contain the highly conserved high mobility group box (HMG-box) region. Increasing studies have shown that TOX is involved in maintaining tumors and promoting T cell exhaustion. In this review, we summarized the biological functions of TOX and its contribution as related to lymphocytic malignancies. We also discussed the potential role of TOX as an immune biomarker and target in immunotherapy for hematological malignancies. Keywords: Thymocyte selection-associated HMG BOX, Biological function, T cell exhaustion, Lymphocytic malignancies, Immune biomarker, Immunotherapy Background located on different chromosomes and have different TOX (thymocyte selection-associated HMG BOX), a functions (Fig. 1, Table 1)[16]. transcription factor, belongs to the high mobility group box (HMG-box) superfamily. In 2002, Wilkinson et al. TOX demonstrated for the first time that TOX plays an im- TOX, also known as KIAA0808, is located on q12.1 of portant role in the differentiation of CD4 + CD8+ human chromosome 8 and has one transcript. The double-positive thymocytes by gene microarray technol- length of the TOX mRNA is 4076 bp, and it contains ogy. Numerous studies have shown that TOX plays a nine exons that translate into a protein consisting of 526 vital role in the development and formation of various amino acids.
    [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]
  • Transcriptional Control of Tissue-Resident Memory T Cell Generation
    Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2019 © 2019 Filip Cvetkovski All rights reserved ABSTRACT Transcriptional control of tissue-resident memory T cell generation Filip Cvetkovski Tissue-resident memory T cells (TRM) are a non-circulating subset of memory that are maintained at sites of pathogen entry and mediate optimal protection against reinfection. Lung TRM can be generated in response to respiratory infection or vaccination, however, the molecular pathways involved in CD4+TRM establishment have not been defined. Here, we performed transcriptional profiling of influenza-specific lung CD4+TRM following influenza infection to identify pathways implicated in CD4+TRM generation and homeostasis. Lung CD4+TRM displayed a unique transcriptional profile distinct from spleen memory, including up-regulation of a gene network induced by the transcription factor IRF4, a known regulator of effector T cell differentiation. In addition, the gene expression profile of lung CD4+TRM was enriched in gene sets previously described in tissue-resident regulatory T cells. Up-regulation of immunomodulatory molecules such as CTLA-4, PD-1, and ICOS, suggested a potential regulatory role for CD4+TRM in tissues. Using loss-of-function genetic experiments in mice, we demonstrate that IRF4 is required for the generation of lung-localized pathogen-specific effector CD4+T cells during acute influenza infection. Influenza-specific IRF4−/− T cells failed to fully express CD44, and maintained high levels of CD62L compared to wild type, suggesting a defect in complete differentiation into lung-tropic effector T cells.
    [Show full text]
  • Genetic and Genomic Analysis of Hyperlipidemia, Obesity and Diabetes Using (C57BL/6J × TALLYHO/Jngj) F2 Mice
    University of Tennessee, Knoxville TRACE: Tennessee Research and Creative Exchange Nutrition Publications and Other Works Nutrition 12-19-2010 Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P. Stewart Marshall University Hyoung Y. Kim University of Tennessee - Knoxville, [email protected] Arnold M. Saxton University of Tennessee - Knoxville, [email protected] Jung H. Kim Marshall University Follow this and additional works at: https://trace.tennessee.edu/utk_nutrpubs Part of the Animal Sciences Commons, and the Nutrition Commons Recommended Citation BMC Genomics 2010, 11:713 doi:10.1186/1471-2164-11-713 This Article is brought to you for free and open access by the Nutrition at TRACE: Tennessee Research and Creative Exchange. It has been accepted for inclusion in Nutrition Publications and Other Works by an authorized administrator of TRACE: Tennessee Research and Creative Exchange. For more information, please contact [email protected]. Stewart et al. BMC Genomics 2010, 11:713 http://www.biomedcentral.com/1471-2164/11/713 RESEARCH ARTICLE Open Access Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P Stewart1, Hyoung Yon Kim2, Arnold M Saxton3, Jung Han Kim1* Abstract Background: Type 2 diabetes (T2D) is the most common form of diabetes in humans and is closely associated with dyslipidemia and obesity that magnifies the mortality and morbidity related to T2D. The genetic contribution to human T2D and related metabolic disorders is evident, and mostly follows polygenic inheritance. The TALLYHO/ JngJ (TH) mice are a polygenic model for T2D characterized by obesity, hyperinsulinemia, impaired glucose uptake and tolerance, hyperlipidemia, and hyperglycemia.
    [Show full text]
  • Supplementary Table S5. Differentially Expressed Gene Lists of PD-1High CD39+ CD8 Tils According to 4-1BB Expression Compared to PD-1+ CD39- CD8 Tils
    BMJ Publishing Group Limited (BMJ) disclaims all liability and responsibility arising from any reliance Supplemental material placed on this supplemental material which has been supplied by the author(s) J Immunother Cancer Supplementary Table S5. Differentially expressed gene lists of PD-1high CD39+ CD8 TILs according to 4-1BB expression compared to PD-1+ CD39- CD8 TILs Up- or down- regulated genes in Up- or down- regulated genes Up- or down- regulated genes only PD-1high CD39+ CD8 TILs only in 4-1BBneg PD-1high CD39+ in 4-1BBpos PD-1high CD39+ CD8 compared to PD-1+ CD39- CD8 CD8 TILs compared to PD-1+ TILs compared to PD-1+ CD39- TILs CD39- CD8 TILs CD8 TILs IL7R KLRG1 TNFSF4 ENTPD1 DHRS3 LEF1 ITGA5 MKI67 PZP KLF3 RYR2 SIK1B ANK3 LYST PPP1R3B ETV1 ADAM28 H2AC13 CCR7 GFOD1 RASGRP2 ITGAX MAST4 RAD51AP1 MYO1E CLCF1 NEBL S1PR5 VCL MPP7 MS4A6A PHLDB1 GFPT2 TNF RPL3 SPRY4 VCAM1 B4GALT5 TIPARP TNS3 PDCD1 POLQ AKAP5 IL6ST LY9 PLXND1 PLEKHA1 NEU1 DGKH SPRY2 PLEKHG3 IKZF4 MTX3 PARK7 ATP8B4 SYT11 PTGER4 SORL1 RAB11FIP5 BRCA1 MAP4K3 NCR1 CCR4 S1PR1 PDE8A IFIT2 EPHA4 ARHGEF12 PAICS PELI2 LAT2 GPRASP1 TTN RPLP0 IL4I1 AUTS2 RPS3 CDCA3 NHS LONRF2 CDC42EP3 SLCO3A1 RRM2 ADAMTSL4 INPP5F ARHGAP31 ESCO2 ADRB2 CSF1 WDHD1 GOLIM4 CDK5RAP1 CD69 GLUL HJURP SHC4 GNLY TTC9 HELLS DPP4 IL23A PITPNC1 TOX ARHGEF9 EXO1 SLC4A4 CKAP4 CARMIL3 NHSL2 DZIP3 GINS1 FUT8 UBASH3B CDCA5 PDE7B SOGA1 CDC45 NR3C2 TRIB1 KIF14 TRAF5 LIMS1 PPP1R2C TNFRSF9 KLRC2 POLA1 CD80 ATP10D CDCA8 SETD7 IER2 PATL2 CCDC141 CD84 HSPA6 CYB561 MPHOSPH9 CLSPN KLRC1 PTMS SCML4 ZBTB10 CCL3 CA5B PIP5K1B WNT9A CCNH GEM IL18RAP GGH SARDH B3GNT7 C13orf46 SBF2 IKZF3 ZMAT1 TCF7 NECTIN1 H3C7 FOS PAG1 HECA SLC4A10 SLC35G2 PER1 P2RY1 NFKBIA WDR76 PLAUR KDM1A H1-5 TSHZ2 FAM102B HMMR GPR132 CCRL2 PARP8 A2M ST8SIA1 NUF2 IL5RA RBPMS UBE2T USP53 EEF1A1 PLAC8 LGR6 TMEM123 NEK2 SNAP47 PTGIS SH2B3 P2RY8 S100PBP PLEKHA7 CLNK CRIM1 MGAT5 YBX3 TP53INP1 DTL CFH FEZ1 MYB FRMD4B TSPAN5 STIL ITGA2 GOLGA6L10 MYBL2 AHI1 CAND2 GZMB RBPJ PELI1 HSPA1B KCNK5 GOLGA6L9 TICRR TPRG1 UBE2C AURKA Leem G, et al.
    [Show full text]
  • Serum Albumin OS=Homo Sapiens
    Protein Name Cluster of Glial fibrillary acidic protein OS=Homo sapiens GN=GFAP PE=1 SV=1 (P14136) Serum albumin OS=Homo sapiens GN=ALB PE=1 SV=2 Cluster of Isoform 3 of Plectin OS=Homo sapiens GN=PLEC (Q15149-3) Cluster of Hemoglobin subunit beta OS=Homo sapiens GN=HBB PE=1 SV=2 (P68871) Vimentin OS=Homo sapiens GN=VIM PE=1 SV=4 Cluster of Tubulin beta-3 chain OS=Homo sapiens GN=TUBB3 PE=1 SV=2 (Q13509) Cluster of Actin, cytoplasmic 1 OS=Homo sapiens GN=ACTB PE=1 SV=1 (P60709) Cluster of Tubulin alpha-1B chain OS=Homo sapiens GN=TUBA1B PE=1 SV=1 (P68363) Cluster of Isoform 2 of Spectrin alpha chain, non-erythrocytic 1 OS=Homo sapiens GN=SPTAN1 (Q13813-2) Hemoglobin subunit alpha OS=Homo sapiens GN=HBA1 PE=1 SV=2 Cluster of Spectrin beta chain, non-erythrocytic 1 OS=Homo sapiens GN=SPTBN1 PE=1 SV=2 (Q01082) Cluster of Pyruvate kinase isozymes M1/M2 OS=Homo sapiens GN=PKM PE=1 SV=4 (P14618) Glyceraldehyde-3-phosphate dehydrogenase OS=Homo sapiens GN=GAPDH PE=1 SV=3 Clathrin heavy chain 1 OS=Homo sapiens GN=CLTC PE=1 SV=5 Filamin-A OS=Homo sapiens GN=FLNA PE=1 SV=4 Cytoplasmic dynein 1 heavy chain 1 OS=Homo sapiens GN=DYNC1H1 PE=1 SV=5 Cluster of ATPase, Na+/K+ transporting, alpha 2 (+) polypeptide OS=Homo sapiens GN=ATP1A2 PE=3 SV=1 (B1AKY9) Fibrinogen beta chain OS=Homo sapiens GN=FGB PE=1 SV=2 Fibrinogen alpha chain OS=Homo sapiens GN=FGA PE=1 SV=2 Dihydropyrimidinase-related protein 2 OS=Homo sapiens GN=DPYSL2 PE=1 SV=1 Cluster of Alpha-actinin-1 OS=Homo sapiens GN=ACTN1 PE=1 SV=2 (P12814) 60 kDa heat shock protein, mitochondrial OS=Homo
    [Show full text]
  • Large Meta-Analysis of Genome-Wide Association Studies
    medRxiv preprint doi: https://doi.org/10.1101/2020.10.01.20200659; this version posted October 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-NC-ND 4.0 International license . Large meta-analysis of genome-wide association studies expands knowledge of the genetic etiology of Alzheimer’s disease and highlights potential translational opportunities Céline Bellenguez1,*,#, Fahri Küçükali2,3,4*, Iris Jansen5,6*, Victor Andrade7,8*, Sonia Morenau- Grau9,10,*, Najaf Amin11,12, Benjamin Grenier-Boley1, Anne Boland13, Luca Kleineidam7,8, Peter Holmans14, Pablo Garcia9,10, Rafael Campos Martin7, Adam Naj15,16, Yang Qiong17, Joshua C. Bis18, Vincent Damotte1, Sven Van der Lee5,6,19, Marcos Costa1, Julien Chapuis1, Vilmentas Giedraitis20, María Jesús Bullido10,21, Adolfo López de Munáin10,22, Jordi Pérez- Tur10,23, Pascual Sánchez-Juan10,24, Raquel Sánchez-Valle25, Victoria Álvarez26, Pau Pastor27, Miguel Medina10,28, Jasper Van Dongen2,3,4, Christine Van Broeckhoven2,3,4, Rik Vandenberghe29,30, Sebastiaan Engelborghs31,32, Gael Nicolas33, Florence Pasquier34, Olivier Hanon35, Carole Dufouil36, Claudine Berr37, Stéphanie Debette36, Jean-François Dartigues36, Gianfranco Spalletta38, Benedetta Nacmias39,40, Vincenzo Solfrezzi41, Barbara Borroni42, Lucio Tremolizzo43, Davide Seripa44, Paolo Caffarra45, Antonio Daniele46,47, Daniela Galimberti48,49, Innocenzo Rainero50, Luisa Benussi51, Alesio Squassina52, Patrizia Mecoci53, Lucilla Parnetti54, Carlo Masullo55, Beatrice Arosio56, John Hardy57, Simon Mead58, Kevin Morgan59, Clive Holmes60, Patrick Kehoe61, Bob Woods62, EADB, Charge, ADGC, Jin Sha15,16, Yi Zhao15,63, Chien-Yueh Lee15,63, Pavel P.
    [Show full text]
  • Meta-Analysis of Genetic Association with Diagnosed Alzheimer's Disease Identifies Novel Risk Loci and Implicates Abeta, Tau, Immunity and Lipid Processing
    bioRxiv preprint doi: https://doi.org/10.1101/294629; this version posted April 4, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Meta-analysis of genetic association with diagnosed Alzheimer's disease identifies novel risk loci and implicates Abeta, Tau, immunity and lipid processing Kunkle BW+*1, Grenier-Boley B+2,3,4, Sims R5, Bis JC6, Naj AC7, Boland A8, Vronskaya M5, van der Lee SJ9, Amlie- Wolf A10, Bellenguez C2,3,4, Frizatti A5, Chouraki V2,11, Martin ER1,12, Sleegers K13,14, Badarinarayan N5, Jakobsdottir J15, Hamilton-Nelson KL1, Aloso R8, Raybould R5, Chen Y10, Kuzma AB10, Hiltunen M17,18, Morgan T5, Ahmad S9, Vardarajan BN19-21, Epelbaum J22, Hoffmann P23,24,25, Boada M26, Beecham GW1,12, Garnier JG8, Harold D27, Fitzpatrick AL28,29, Valladares O10, Moutet ML8, Gerrish A5, Smith AV30,31, Qu L10, Bacq D8, Denning N5, Jian X32, Zhao Y10, Zompo MD33, Fox NC34, Grove ML23, Choi SH16, Mateo I35, Hughes JT36, Adams HH9, Malamon J10, Garcia FS36, Patel Y37, Brody JA6, Dombroski B10, Naranjo MCD36, Daniilidou M38, Eiriksdottir G15, Mukherjee S39, Wallon D40,41, Uphill J42, Aspelund T15,43, Cantwell LB10, Garzia F8, Galimberti D44, Hofer E45,46, Butkiewics M47, Fin B8, Scarpini E44, Sarnowski C16, Bush W47, Meslage S8, Kornhuber J48, White CC49, Song Y47, Barber RC50, Engelborghs S51,52, Pichler S53, Voijnovic D9, Adams PM54, Vandenberghe
    [Show full text]
  • List of Down-Regulated Transcripts
    Supplementary Table 1: List of down-regulated transcripts (Fold Change>1.7 and p<0.05). List of the transcipts downregulated by Tpr-Met in P27 cachectic gastrocnemius muscles. Shown are the transcripts gene annotation: Mus Musculus Entrez Gene ID, official symbol from NCBI, average (AVG) Log2 Ratio derived from Illumina microarrays (3 replicates/condition), fold change and p-value computations, Log2 ratio over AVG Log2 controls and full definition of the genes. All genes modulated with a fold change > 1.7 and p-value<0.05 are included. AVG Entrez Gene Fold Symbol Log2 T-test Ctrl Ctrl Ctrl TM TM TM Definition ID Change Ratio - - - - 26908 Eif2s3y 3.598 12.1 0.000 0.146 0.248 0.102 3.712 3.571 -3.510 eukaryotic translation initiation factor 2, subunit 3, structural gene Y-linked (Eif2s3y), mRNA. - - - 13628 Eef1a2 1.333 2.5 0.011 -0.444 0.261 0.183 1.503 0.950 -1.546 eukaryotic translation elongation factor 1 alpha 2 (Eef1a2), mRNA. - - - 13628 Eef1a2 1.298 2.5 0.031 -0.383 0.275 0.108 1.632 0.605 -1.656 eukaryotic translation elongation factor 1 alpha 2 (Eef1a2), mRNA. - - - 56222 Cited4 1.268 2.4 0.029 -0.466 0.035 0.431 1.109 0.889 -1.805 Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain, 4 (Cited4), mRNA. - - - 18810 Plec1 1.246 2.4 0.022 -0.440 0.327 0.113 1.303 0.779 -1.657 plectin 1 (Plec1), transcript variant 10, mRNA. - - - 20741 Spnb1 1.238 2.4 0.016 -0.384 0.314 0.071 1.129 0.902 -1.684 spectrin beta 1 (Spnb1), mRNA.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Dynamic Gene Expression by Putative Hair-Cell Progenitors During
    Dynamic gene expression by putative hair-cell PNAS PLUS progenitors during regeneration in the zebrafish lateral line Aaron B. Steiner1, Taeryn Kim, Victoria Cabot2, and A. J. Hudspeth Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY 10065 Edited* by Yuh Nung Jan, Howard Hughes Medical Institute, University of California, San Francisco, CA, and approved February 25, 2014 (received for review October 2, 2013) Hearing loss is most commonly caused by the destruction of been identified, however, and even fewer have been confirmed as mechanosensory hair cells in the ear. This condition is usually mantle cell-specific (15–17). permanent: Despite the presence of putative hair-cell progenitors Although previous transcriptomic screens have sought genes in the cochlea, hair cells are not naturally replenished in adult expressed in the lateral line, none has focused on mantle cells mammals. Unlike those of the mammalian ear, the progenitor cells (18–20). The results of such studies reflect gene expression in of nonmammalian vertebrates can regenerate hair cells through- several cell types, a complication that might mask gene expres- out life. The basis of this difference remains largely unexplored sion in progenitors. One factor impeding the isolation and char- but may lie in molecular dissimilarities that affect how progenitors acterization of progenitor cells has been the lack of a transgenic respond to hair-cell death. To approach this issue, we analyzed line in which these cells are inclusively and specifically labeled, gene expression in hair-cell progenitors of the lateral-line system. allowing their separation by cell sorting.
    [Show full text]