Genome Architecture and Transcription Data Reveal Allelic Bias During the Cell Cycle

Total Page:16

File Type:pdf, Size:1020Kb

Genome Architecture and Transcription Data Reveal Allelic Bias During the Cell Cycle bioRxiv preprint doi: https://doi.org/10.1101/2020.03.15.992164; this version posted April 21, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. Genome architecture and transcription data reveal allelic bias during the cell cycle Stephen Lindsly1, Wenlong Jia2, Haiming Chen1, Sijia Liu3, Scott Ronquist1, Can Chen4;7, Xingzhao Wen5, Gilbert Omenn1, Shuai Cheng Li2, Max Wicha1;6, Alnawaz Rehemtulla6, Lindsey Muir1, Indika Rajapakse1;7;∗ 1Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor 2Department of Computer Science, City University of Hong Kong 3MIT-IBM Watson AI Lab, IBM Research 4Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor 5Department of Bioengineering, University of California San Diego, La Jolla 6Department of Hematology/Oncology, University of Michigan, Ann Arbor 7Department of Mathematics, University of Michigan, Ann Arbor ∗To whom correspondence should be addressed; E-mail: [email protected]. bioRxiv preprint doi: https://doi.org/10.1101/2020.03.15.992164; this version posted April 21, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. Highlights • Structural and functional differences between the maternal and paternal genomes, includ- ing similar allelic bias in related subsets of genes. • Coupling between the dynamics of gene expression and genome architecture for specific alleles illuminates an allele-specific 4D Nucleome. • Introduction of a novel allele-specific phasing algorithm for genome architecture and a quantitative framework for integration of gene expression and genome architecture. Abstract Millions of genetic variants exist between paternal and maternal genomes in human cells, which result in unequal allelic contributions to gene transcrip- tion. We sought to understand differences in transcription and genome archi- tecture between the maternal and paternal genomes across the cell cycle. Our approach integrates haplotype-resolved genome-wide data from B-lymphocytes in G1, S, and G2/M, mature and nascent mRNA, genome wide chromosome conformation capture (Hi-C), and protein binding (ChIP-seq). Further, our novel method, HaploHiC, enables phasing of reads of unknown origin. Our data indicate significant allelic bias in cell identity genes, and notably absent bias in universally essential cell cycle and circadian rhythm genes. We offer a model in which coordinated biallelic expression reflects prioritized preserva- tion of essential gene sets. bioRxiv preprint doi: https://doi.org/10.1101/2020.03.15.992164; this version posted April 21, 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. Introduction Maternal and paternal chromosomes typically each encode an allele of the same gene. Mil- lions of single nucleotide variants (SNVs), insertions, and deletions (InDels) enable distinction between the maternal and paternal genome, and DNA sequence variations can influence 3D conformation of the genome (1). Studying the maternal and paternal alleles of a gene may en- hance our understanding of genome organization and mechanisms of bias in allele expression. Allelic differences have been observed through gene expression, 3D chromatin organization, and protein binding (2–9), and it has been shown that even subtle differences in genome struc- ture can lead to differential gene expression and regulation (10). Approximately 10-25% of all genes have allelic biased expression across many cell types (11–13). One well studied example of allele-specific expression and architecture is X chromosome inactivation (XCI) in females, in which one of the two X chromosomes is tightly packed into two ‘superdomains’ with silencing of gene expression (4, 14). Another example of known allele-specific expression is imprinting, in which one allele’s expression is suppressed (15). It has also been shown for single cells that monoallelic transcripts could make up over 75% of total transcripts (16). The allele-specific nature of genome architecture and gene expression have been explored individually, but it remains poorly understood how differences in chromosome architecture be- tween alleles contribute to differences in gene expression. To address this problem, we look to transcription factors and other regulatory proteins as key components that connect differences in genome conformation with differential expression of the maternal and paternal genomes. By integrating RNA sequencing (RNA-seq), bromouridine sequencing (Bru-seq), and genome- wide chromosome conformation capture (Hi-C), along with publicly available protein binding data (ChIP-seq), we contribute insight into how maternal and paternal genomes contribute to cell phenotype. The redundancy inherent in diploid genomes makes them robust to mutations and genetic loss-of-function events. In cancers, this redundancy is often missing, and referred to as loss of heterozygosity. For example, BRCA1 and BRCA2 exhibit loss of heterozygosity (17), and bioRxiv preprint doi: https://doi.org/10.1101/2020.03.15.992164; this version posted April 21, 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. mutations in these genes lead to a significant increase in the likelihood of developing breast cancer (18). Additionally, monoallelically expressed genes, like X-inactivated and imprinted genes (15, 19–21), do not have a ‘backup’ gene to compensate for potential deleterious muta- tions, and it has been found that mutations in monoallelicly expressed genes are responsible for disease phenotypes (22). These findings lend credence to the pursuit of allele-specific genome analysis. The 4D Nucleome (4DN) describes the relationship between genome architecture, gene expression, and cell phenotype through time (23). Chen et al. (24) introduced a mathematical framework for the 4DN, which can be applied in an allele-specific manner to a fully haplotyped cell line (e.g. NA12878). Despite extensive study of genome replication and compaction in cell division, few studies focus on differences between maternal and paternal genomes during these processes. For example, the two alleles may have different dynamics through the cell cycle that influence proliferation, or an allele-specific 4DN. In this paper, we integrate allele-specific RNA-seq, Bru-seq, Hi-C (Figure 1), and ChIP-seq data to provide a more comprehensive understanding of the maternal and paternal contributions to phenotype, and the relationship between these properties. We analyze differential expression for allele-biases and allele-specific cell cycle-biases, chromatin compartment switching (also known as A/B compartments), and Topologically Associated Domains (TADs). In addition, we analyze our Hi-C data using our recently developed methods that help discern differences in genome architecture between the two genomes and through the cell cycle. Our methods for analyses reveal notable patterns of allelic bias across the cell cycle, including subsets of genes involved in related cellular processes. This work supports the idea that separate analysis of the two parental genomes is imperative to future genomic studies. bioRxiv preprint doi: https://doi.org/10.1101/2020.03.15.992164; this version posted April 21, 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. Fig. 1: Experimental and allelic separation workflow. Cell cycle sorted cells were extracted for RNA-seq, Bru-seq, and Hi-C (left to right, respectively). RNA-seq and Bru-seq data were allelicly phased via SNVs/InDels (left). SNV/InDel based imputation and allelic phasing done using HaploHiC (right). bioRxiv preprint doi: https://doi.org/10.1101/2020.03.15.992164; this version posted April 21, 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. Results Chromosome level differences of the maternal and paternal genomes In order to establish whether there are allele-specific differences in both chromosome structure and gene expression, we analyzed parentally phased whole-chromosome Hi-C matrices and RNA-seq data at a 1 Mb scale (phasing discussed below, in Supplementary Notes 2.1.7, and 2.4). We performed a simple subtraction of the phased Hi-C matrices and found the Frobenius norm of the difference matrices, adjusted for chromosome size. Similarly, we subtracted the phased RNA-seq vectors and found the Frobenius norm of each difference vector, adjusted for chromosome size. If the two parents’ genomes were equivalent, the resulting chromosome-level Frobenius norms would approach zero. As seen in Figure 2, we found that all chromosomes have allelic differences in both structure (blue) and function (red). Unsurprisingly, chromosome X had the largest structural difference, followed by chromosomes 9, 21, and 14. The X chro- mosome also had the most extreme functional difference, followed by chromosomes 13, 7, and 9. We standardized the Frobenius norms of all chromosomes by the norm of the X chromo- some, as its value was much larger than any of the autosomes.
Recommended publications
  • Downloaded from [266]
    Patterns of DNA methylation on the human X chromosome and use in analyzing X-chromosome inactivation by Allison Marie Cotton B.Sc., The University of Guelph, 2005 A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY in The Faculty of Graduate Studies (Medical Genetics) THE UNIVERSITY OF BRITISH COLUMBIA (Vancouver) January 2012 © Allison Marie Cotton, 2012 Abstract The process of X-chromosome inactivation achieves dosage compensation between mammalian males and females. In females one X chromosome is transcriptionally silenced through a variety of epigenetic modifications including DNA methylation. Most X-linked genes are subject to X-chromosome inactivation and only expressed from the active X chromosome. On the inactive X chromosome, the CpG island promoters of genes subject to X-chromosome inactivation are methylated in their promoter regions, while genes which escape from X- chromosome inactivation have unmethylated CpG island promoters on both the active and inactive X chromosomes. The first objective of this thesis was to determine if the DNA methylation of CpG island promoters could be used to accurately predict X chromosome inactivation status. The second objective was to use DNA methylation to predict X-chromosome inactivation status in a variety of tissues. A comparison of blood, muscle, kidney and neural tissues revealed tissue-specific X-chromosome inactivation, in which 12% of genes escaped from X-chromosome inactivation in some, but not all, tissues. X-linked DNA methylation analysis of placental tissues predicted four times higher escape from X-chromosome inactivation than in any other tissue. Despite the hypomethylation of repetitive elements on both the X chromosome and the autosomes, no changes were detected in the frequency or intensity of placental Cot-1 holes.
    [Show full text]
  • A Worldwide Map of Swine Short Tandem Repeats and Their
    Wu et al. Genet Sel Evol (2021) 53:39 https://doi.org/10.1186/s12711-021-00631-4 Genetics Selection Evolution RESEARCH ARTICLE Open Access A worldwide map of swine short tandem repeats and their associations with evolutionary and environmental adaptations Zhongzi Wu1, Huanfa Gong1, Mingpeng Zhang1, Xinkai Tong1, Huashui Ai1, Shijun Xiao1, Miguel Perez‑Enciso2,3, Bin Yang1* and Lusheng Huang1* Abstract Background: Short tandem repeats (STRs) are genetic markers with a greater mutation rate than single nucleotide polymorphisms (SNPs) and are widely used in genetic studies and forensics. However, most studies in pigs have focused only on SNPs or on a limited number of STRs. Results: This study screened 394 deep‑sequenced genomes from 22 domesticated pig breeds/populations world‑ wide, wild boars from both Europe and Asia, and numerous outgroup Suidaes, and identifed a set of 878,967 poly‑ morphic STRs (pSTRs), which represents the largest repository of pSTRs in pigs to date. We found multiple lines of evidence that pSTRs in coding regions were afected by purifying selection. The enrichment of trinucleotide pSTRs in coding sequences (CDS), 5′UTR and H3K4me3 regions suggests that trinucleotide STRs serve as important com‑ ponents in the exons and promoters of the corresponding genes. We demonstrated that, compared to SNPs, pSTRs provide comparable or even greater accuracy in determining the breed identity of individuals. We identifed pSTRs that showed signifcant population diferentiation between domestic pigs and wild boars in Asia and Europe. We also observed that some pSTRs were signifcantly associated with environmental variables, such as average annual tem‑ perature or altitude of the originating sites of Chinese indigenous breeds, among which we identifed loss‑of‑function and/or expanded STRs overlapping with genes such as AHR, LAS1L and PDK1.
    [Show full text]
  • A High-Throughput Approach to Uncover Novel Roles of APOBEC2, a Functional Orphan of the AID/APOBEC Family
    Rockefeller University Digital Commons @ RU Student Theses and Dissertations 2018 A High-Throughput Approach to Uncover Novel Roles of APOBEC2, a Functional Orphan of the AID/APOBEC Family Linda Molla Follow this and additional works at: https://digitalcommons.rockefeller.edu/ student_theses_and_dissertations Part of the Life Sciences Commons A HIGH-THROUGHPUT APPROACH TO UNCOVER NOVEL ROLES OF APOBEC2, A FUNCTIONAL ORPHAN OF THE AID/APOBEC FAMILY A Thesis Presented to the Faculty of The Rockefeller University in Partial Fulfillment of the Requirements for the degree of Doctor of Philosophy by Linda Molla June 2018 © Copyright by Linda Molla 2018 A HIGH-THROUGHPUT APPROACH TO UNCOVER NOVEL ROLES OF APOBEC2, A FUNCTIONAL ORPHAN OF THE AID/APOBEC FAMILY Linda Molla, Ph.D. The Rockefeller University 2018 APOBEC2 is a member of the AID/APOBEC cytidine deaminase family of proteins. Unlike most of AID/APOBEC, however, APOBEC2’s function remains elusive. Previous research has implicated APOBEC2 in diverse organisms and cellular processes such as muscle biology (in Mus musculus), regeneration (in Danio rerio), and development (in Xenopus laevis). APOBEC2 has also been implicated in cancer. However the enzymatic activity, substrate or physiological target(s) of APOBEC2 are unknown. For this thesis, I have combined Next Generation Sequencing (NGS) techniques with state-of-the-art molecular biology to determine the physiological targets of APOBEC2. Using a cell culture muscle differentiation system, and RNA sequencing (RNA-Seq) by polyA capture, I demonstrated that unlike the AID/APOBEC family member APOBEC1, APOBEC2 is not an RNA editor. Using the same system combined with enhanced Reduced Representation Bisulfite Sequencing (eRRBS) analyses I showed that, unlike the AID/APOBEC family member AID, APOBEC2 does not act as a 5-methyl-C deaminase.
    [Show full text]
  • Genome Architecture and Transcription Data Reveal Allelic Bias During the Cell Cycle
    bioRxiv preprint doi: https://doi.org/10.1101/2020.03.15.992164; this version posted April 1, 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. Genome architecture and transcription data reveal allelic bias during the cell cycle Stephen Lindsly1, Wenlong Jia2, Haiming Chen1, Sijia Liu3, Scott Ronquist1, Can Chen4;7, Xingzhao Wen5, Gilbert Omenn1, Shuai Cheng Li2, Max Wicha1;6, Alnawaz Rehemtulla6, Lindsey Muir1, Indika Rajapakse1;7;∗ 1Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor 2Department of Computer Science, City University of Hong Kong 3MIT-IBM Watson AI Lab, IBM Research 4Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor 5Department of Bioengineering, University of California San Diego, La Jolla 6Department of Hematology/Oncology, University of Michigan, Ann Arbor 7Department of Mathematics, University of Michigan, Ann Arbor ∗To whom correspondence should be addressed; E-mail: [email protected]. bioRxiv preprint doi: https://doi.org/10.1101/2020.03.15.992164; this version posted April 1, 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. Highlights • Structural and functional differences between the maternal and paternal genomes, includ- ing similar allelic bias in related subsets of genes. • Coupling between the dynamics of gene expression and genome architecture for specific alleles illuminates an allele-specific 4D Nucleome. • Introduction of a novel allele-specific phasing algorithm for genome architecture and a quantitative framework for integration of gene expression and genome architecture.
    [Show full text]
  • A Temporally Controlled Sequence of X-Chromosome Inactivation and Reactivation Defines Female Mouse in Vitro Germ Cells with Meiotic Potential
    bioRxiv preprint doi: https://doi.org/10.1101/2021.08.11.455976; this version posted August 11, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. A temporally controlled sequence of X-chromosome inactivation and reactivation defines female mouse in vitro germ cells with meiotic potential Jacqueline Severino1†, Moritz Bauer1,9†, Tom Mattimoe1, Niccolò Arecco1, Luca Cozzuto1, Patricia Lorden2, Norio Hamada3, Yoshiaki Nosaka4,5,6, So Nagaoka4,5,6, Holger Heyn2, Katsuhiko Hayashi7, Mitinori Saitou4,5,6 and Bernhard Payer1,8* Abstract The early mammalian germ cell lineage is characterized by extensive epigenetic reprogramming, which is required for the maturation into functional eggs and sperm. In particular, the epigenome needs to be reset before parental marks can be established and then transmitted to the next generation. In the female germ line, reactivation of the inactive X- chromosome is one of the most prominent epigenetic reprogramming events, and despite its scale involving an entire chromosome affecting hundreds of genes, very little is known about its kinetics and biological function. Here we investigate X-chromosome inactivation and reactivation dynamics by employing a tailor-made in vitro system to visualize the X-status during differentiation of primordial germ cell-like cells (PGCLCs) from female mouse embryonic stem cells (ESCs). We find that the degree of X-inactivation in PGCLCs is moderate when compared to somatic cells and characterized by a large number of genes escaping full inactivation.
    [Show full text]
  • The Chromatin Associated Phf12 Protein Maintains Nucleolar Integrity And
    MCB Accepted Manuscript Posted Online 12 December 2016 Mol. Cell. Biol. doi:10.1128/MCB.00522-16 Copyright © 2016, American Society for Microbiology. All Rights Reserved. 1 The chromatin associated Phf12 protein maintains nucleolar integrity and 2 prevents premature cellular senescence. Downloaded from 3 4 Running Title: Pf1 regulates ribosomal biogenesis 5 6 Richard Graveline 1, #, Katarzyna Marcinkiewicz 1, #, Seyun Choi 1, Marilène Paquet 2, http://mcb.asm.org/ 7 Wolfgang Wurst 3, Thomas Floss 3, and Gregory David 1, * 8 1. New York University School of Medicine, New York, NY, United States. 9 2. Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe (Québec), 10 Canada 11 3. Helmholtz Zentrum München - German Research Center for Environmental Health, on February 2, 2017 by GSF Forschungszentrum F 12 Neuherberg, Germany 13 # Contributed equally 14 *CORRESPONDENCE: Gregory David 15 Dept. of Biochemistry and Molecular Pharmacology, MSB417 16 NYU Langone Medical Center 17 550 First Ave. 18 New York, NY10016. 19 [email protected] 20 Ph: 212-263-2926; Fax: 212-263-7133 21 22 23 KEYWORDS: Transcription; Ribosome; Pf1; Senescence; Nucleolus 24 25 Abstract 26 PF1, also known as PHF12 (plant homeodomain (PHD) zinc finger protein 12) is a member 27 of the PHD zinc finger family of proteins. PF1 associates with a chromatin interacting Downloaded from 28 protein complex comprised of MRG15, Sin3B, and HDAC1, that functions as a 29 transcriptional modulator. The biological function of Pf1 remains largely elusive. We 30 undertook the generation of Pf1 knockout mice to elucidate its physiological role. We 31 demonstrate that Pf1 is required for mid-to-late gestation viability.
    [Show full text]
  • Dynamic Erasure of Random X-Chromosome Inactivation During Ipsc Reprogramming
    bioRxiv preprint doi: https://doi.org/10.1101/545558; this version posted February 9, 2019. 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 4.0 International license. Dynamic Erasure of Random X-Chromosome Inactivation during iPSC Reprogramming Adrian Janiszewski1,*, Irene Talon1,*, Juan Song1, Natalie De Geest1, San Kit To1, Greet Bervoets2,3, Jean-Christophe Marine2,3, Florian Rambow2,3, Vincent Pasque1,✉. 1 KU Leuven - University of Leuven, Department of Development and Regeneration, Herestraat 49, B-3000 Leuven, Belgium. 2 Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology 3 Department of Oncology, KU Leuven, Belgium ✉ Correspondence to: [email protected] (V.P.) * These authors contributed equally Format: Research paper ABSTRACT Background: Induction and reversal of chromatin silencing is critical for successful development, tissue homeostasis and the derivation of induced pluripotent stem cells (iPSCs). X-chromosome inactivation (XCI) and reactivation (XCR) in female cells represent chromosome-wide transitions between active and inactive chromatin states. While XCI has long been studied and provided important insights into gene regulation, the dynamics and mechanisms underlying the reversal of stable chromatin silencing of X-linked genes are much less understood. Here, we use allele- specific transcriptomic approaches to study XCR during mouse iPSC reprogramming in order to elucidate the timing and mechanisms of chromosome-wide reversal of gene silencing. Results: We show that XCR is hierarchical, with subsets of genes reactivating early, late and very late.
    [Show full text]
  • Paternal X Inactivation Does Not Correlate with X Chromosome Evolutionary Strata in Marsupials Claudia L Rodríguez-Delgado1*, Shafagh a Waters2 and Paul D Waters2*
    Rodríguez-Delgado et al. BMC Evolutionary Biology (2014) 14:267 DOI 10.1186/s12862-014-0267-z RESEARCH ARTICLE Open Access Paternal X inactivation does not correlate with X chromosome evolutionary strata in marsupials Claudia L Rodríguez-Delgado1*, Shafagh A Waters2 and Paul D Waters2* Abstract Background: X chromosome inactivation is the transcriptional silencing of one X chromosome in the somatic cells of female mammals. In eutherian mammals (e.g. humans) one of the two X chromosomes is randomly chosen for silencing, with about 15% (usually in younger evolutionary strata of the X chromosome) of genes escaping this silencing. In contrast, in the distantly related marsupial mammals the paternally derived X is silenced, although not as completely as the eutherian X. A chromosome wide examination of X inactivation, using RNA-seq, was recently undertaken in grey short-tailed opossum (Monodelphis domestica) brain and extraembryonic tissues. However, no such study has been conduced in Australian marsupials, which diverged from their American cousins ~80 million years ago, leaving a large gap in our understanding of marsupial X inactivation. Results: We used RNA-seq data from blood or liver of a family (mother, father and daughter) of tammar wallabies (Macropus eugenii), which in conjunction with available genome sequence from the mother and father, permitted genotyping of 42 expressed heterozygous SNPs on the daughter’s X. These 42 SNPs represented 34 X loci, of which 68% (23 of the 34) were confirmed as inactivated on the paternally derived X in the daughter’sliver; the remaining 11 X loci escaped inactivation. Seven of the wallaby loci sampled were part of the old X evolutionary stratum, of which three escaped inactivation.
    [Show full text]
  • Content Based Search in Gene Expression Databases and a Meta-Analysis of Host Responses to Infection
    Content Based Search in Gene Expression Databases and a Meta-analysis of Host Responses to Infection A Thesis Submitted to the Faculty of Drexel University by Francis X. Bell in partial fulfillment of the requirements for the degree of Doctor of Philosophy November 2015 c Copyright 2015 Francis X. Bell. All Rights Reserved. ii Acknowledgments I would like to acknowledge and thank my advisor, Dr. Ahmet Sacan. Without his advice, support, and patience I would not have been able to accomplish all that I have. I would also like to thank my committee members and the Biomed Faculty that have guided me. I would like to give a special thanks for the members of the bioinformatics lab, in particular the members of the Sacan lab: Rehman Qureshi, Daisy Heng Yang, April Chunyu Zhao, and Yiqian Zhou. Thank you for creating a pleasant and friendly environment in the lab. I give the members of my family my sincerest gratitude for all that they have done for me. I cannot begin to repay my parents for their sacrifices. I am eternally grateful for everything they have done. The support of my sisters and their encouragement gave me the strength to persevere to the end. iii Table of Contents LIST OF TABLES.......................................................................... vii LIST OF FIGURES ........................................................................ xiv ABSTRACT ................................................................................ xvii 1. A BRIEF INTRODUCTION TO GENE EXPRESSION............................. 1 1.1 Central Dogma of Molecular Biology........................................... 1 1.1.1 Basic Transfers .......................................................... 1 1.1.2 Uncommon Transfers ................................................... 3 1.2 Gene Expression ................................................................. 4 1.2.1 Estimating Gene Expression ............................................ 4 1.2.2 DNA Microarrays ......................................................
    [Show full text]
  • Immune Cells in Spleen and Mucosa + CD127 Neg Production of IL-17 and IL-22 by CD3 TLR5 Signaling Stimulates the Innate
    Downloaded from http://www.jimmunol.org/ by guest on September 23, 2021 neg is online at: average * The Journal of Immunology , 23 of which you can access for free at: 2010; 185:1177-1185; Prepublished online 21 Immune Cells in Spleen and Mucosa from submission to initial decision + 4 weeks from acceptance to publication TLR5 Signaling Stimulates the Innate Production of IL-17 and IL-22 by CD3 CD127 Laurye Van Maele, Christophe Carnoy, Delphine Cayet, Pascal Songhet, Laure Dumoutier, Isabel Ferrero, Laure Janot, François Erard, Julie Bertout, Hélène Leger, Florent Sebbane, Arndt Benecke, Jean-Christophe Renauld, Wolf-Dietrich Hardt, Bernhard Ryffel and Jean-Claude Sirard June 2010; doi: 10.4049/jimmunol.1000115 http://www.jimmunol.org/content/185/2/1177 J Immunol cites 51 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://www.jimmunol.org/content/185/2/1177.full#ref-list-1 http://www.jimmunol.org/content/suppl/2010/06/21/jimmunol.100011 5.DC1 This article Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2010 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606.
    [Show full text]
  • Analog-Sensitive Cell Line Identifies Cellular Substrates of CDK9
    www.oncotarget.com Oncotarget, 2019, Vol. 10, (No. 65), pp: 6934-6943 Research Paper Analog-sensitive cell line identifies cellular substrates of CDK9 Tim-Michael Decker1,4, Ignasi Forné2, Tobias Straub3, Hesham Elsaman1, Guoli Ma1, Nilay Shah1,5, Axel Imhof2 and Dirk Eick1 1Department of Molecular Epigenetics, Helmholtz Center Munich and Center for Integrated Protein Science Munich (CIPSM), Germany 2Biomedical Center Munich, ZFP, Ludwig-Maximilian University Munich, Germany 3Bioinformatic Unit, Biomedical Center Munich, Ludwig-Maximilian University Munich, Planegg-Martinsried, Germany 4Present address: Department of Biochemistry, University of Colorado, Boulder, USA 5Present address: Stowers Institute for Medical Research, Kansas City, Missouri, USA Correspondence to: Tim-Michael Decker, email: [email protected] Dirk Eick, email: [email protected] Keywords: CDK9; protein kinase; transcription; RNA splicing; phosphoproteomics Received: September 12, 2019 Accepted: November 07, 2019 Published: December 10, 2019 Copyright: Decker et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Transcriptional cyclin-dependent kinases regulate all phases of transcription. Cyclin-dependent kinase 9 (CDK9) has been implicated in the regulation of promoter- proximal pausing of RNA polymerase II and more recently in transcription termination. Study of the substrates of CDK9 has mostly been limited to in vitro approaches that lack a quantitative assessment of CDK9 activity. Here we analyzed the cellular phosphoproteome upon inhibition of CDK9 by combining analog-sensitive kinase technology with quantitative phosphoproteomics in Raji B-cells.
    [Show full text]
  • Prediction of Pancreatic Adenocarcinoma Patient Risk Status Using Alternative Splicing Events
    Prediction of pancreatic adenocarcinoma patient risk status using alternative splicing events Rajesh Kumar Institute of Microbial Technology CSIR Anjali Lathwal Indraprastha Institute of Information Technology Delhi Gajendra Pal Raghava ( [email protected] ) Indraprastha Institute of Information Technology Delhi https://orcid.org/0000-0002-8902-2876 Research Article Keywords: Alternative Splicing, Pancreatic Adenocarcinoma, Splice Site Mutations, Survival Analysis, Cox Proportional Hazard, Prognostic Biomarker Posted Date: June 3rd, 2021 DOI: https://doi.org/10.21203/rs.3.rs-411576/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/19 Abstract In literature, several mRNA, miRNA, lncRNA based biomarkers are identied by genomic analysis to stratify the patients into high and low risk groups of pancreatic adenocarcinoma (PAAD). The identied biomarkers are of limited use in terms of sensitivity and prediction ability. Thus, we aimed to identify the prognostic alternative splicing events and their related mutations in the PAAD. PAAD splicing data of 174 samples (17874 AS events in 6209 genes) and corresponding clinical information was obtained from the SpliceSeq and The Cancer Genome Atlas (TCGA), respectively. Prognostic-index based modeling was used to obtain the best predictive models for the seven AS types. However, model based on multiple spliced events genes (APP; LATS1; MRPL4; LAS1L; STARD10; PHF21A; NMRAL1) outperformed the single event models with a remarkable HR of 9.13 (p-value = 6.42e-10) as well as other existing models. Results from g:Proler suggest that transcription factors ZF5, ER81, E2F-1/2/3, ER81, Erg, and PEA3 are most related to the prognostic spliced genes.
    [Show full text]