Delineation of the Genome-Wide Recruitment of Hepatitis B Virus Trans-Activator Protein Hbx, in Primary Hepatocytes and Liver Cancer Cells

Total Page:16

File Type:pdf, Size:1020Kb

Delineation of the Genome-Wide Recruitment of Hepatitis B Virus Trans-Activator Protein Hbx, in Primary Hepatocytes and Liver Cancer Cells Delineation of the Genome-wide Recruitment of Hepatitis B Virus Trans-activator Protein HBx, in Primary Hepatocytes and Liver Cancer Cells By Amanda Jayne Lyon B.S. in Biomedical Technology, May 2006, Norwich University M.S. in Public Health Microbiology, Emerging Infectious Diseases, May 2009 The George Washington University A Dissertation submitted to The Faculty of The Columbian College of Arts and Sciences of The George Washington University in partial fulfillment of the requirements for the degree of Doctor of Philosophy August 31, 2012 Dissertation directed by: Rakesh Kumar Professor of Biochemistry and Molecular Biology The Columbian College of Arts and Sciences of The George Washington University certifies that Amanda Jayne Lyon has passed the Final Examination for the degree of Doctor of Philosophy as of August 7, 2012. This is the final and approved form of the dissertation. Delineation of the Genome-wide Recruitment of Hepatitis B Virus Trans-activator Protein HBx, in Primary Hepatocytes and Liver Cancer Cells. Amanda Jayne Lyon Dissertation Research Committee: Rakesh Kumar, Chairman and Professor of Biochemistry and Molecular Biology Paul J. Brindley, Professor of Microbiology, Immunology and Tropical Medicine Ajit Kumar, Professor of Biochemistry and Molecular Biology ii © Copyright “2012” by Amanda Jayne Lyon All Rights Reserved iii Dedication I would like to dedicate this dissertation to my family members; Cherie Lyon, David Lyon, Laurie Lyon, and Shaina Lyon, as well as to my Fiancée, Dirk Vandeveer. You are all instrumental in my life and have always been encouraging and supportive, thank you. iv Acknowledgements I would like to express my gratitude towards everyone who played a role in this experience, to include all of the professors and members of IBS as well as those who contributed to my dissertation research. I would like to thank Dr. Rakesh Kumar and my committee members Dr. Paul Brindley, Dr. Ajit Kumar, Dr. Jeyanthy Eswaran and Dr. Anton Sidawy, for their guidance and support throughout this experience. I am grateful for the opportunity to work on this interesting research project, which has taught me numerous skills that I will take with me. Thank you to all members of Dr. Kumar’s laboratory for your suggestions and guidance. Thank you, Dr. Ali Ramezani, for your help with the lentiviral vectors and Dr. Krishna Banudha for your help with the PHHs. Thank you to everyone who supported me in the completion of this dissertation research project, your assistance is greatly appreciated. I would like to thank Dr. Linda Werling for her support and guidance. Dr. Werling, you are a wonderful person who truly cares for her students and whose leadership in IBS makes the program so successful. Thank you to my Academic Advisor, Dr. David Leitenberg who has always been helpful and supportive. I must thank Dr. Dante Verme who was one of my very first professors at GWU, in the Fall of 2007. You are an outstanding professor and I enjoyed my time as your Graduate Administrative Assistant and Instructional Assistant for GIS. I will miss it. v Thank you to my friends in the IBS, especially the entering class of the Fall of 2009. Thank you to Claire Hoptay, Lindsay Garvin, and Amanda Woerman, who have become close friends of mine. Thank you Ngoc-Han Ha, for your friendship and support throughout this experience, and beyond. Thank you for listening, and for your advice. I will always remember the good times. At the beginning of the Ph.D., I was told that a strong support system is vital to a student’s success. Luckily, I never had to worry about that, and it is due to my amazing family and Fiancée. Mom, you always told me that I could be whatever I wanted to be, and that advice has kept me striving for more. Thank you for lending an open ear when ever I needed to talk. Your support is a vital part of my success. Dad, your support has also been vital to my success. Your words of encouragement and advice often come to my mind. I have always looked up to you and have strived to work as hard, as I know you do. Laurie, “the human dictionary,” you have always been an inspirational sister, who I’ve looked up to and have strived to be like, even though, as someone who is very independent, I would never admit it. Today, I still look up to you and am proud to have you as a role model. Shaina, you are the source of laughter in our family. You make life more fun and enjoyable and I admire that. Thank you for your support and for always being there for me. To the Vandeveers, you have been like family to me, for many years now. Thank you for being so supportive and caring. You are wonderful people who I admire and look up to. Dirk, I could not have completed the M.S., or the Ph.D. without you. Your daily support and encouragement has kept me going, through it all. Our time together has made me a much happier and stronger person. Without you, my academic success would not have been possible, thank you. vi Abstract Delineation of the Genome-wide Recruitment of Hepatitis B Virus Trans-activator Protein HBx, in Primary Hepatocytes and Liver Cancer Cells Hepatocellular carcinoma (HCC) is among the top five cancers worldwide, with Hepatitis B virus (HBV)-associated HCC accounting for the majority of all cases. The cancer promoting activity of HBV is derived from one of its gene products, HBx, the primary trans-activator of cellular genes with roles in carcinogenesis. Although the contribution of HBx to HCC is firmly established, the nature of comparative genome-wide targets of HBx in primary human hepatocytes (PHH) and liver cancer cells, remains unknown, and is being investigated in the present work. Using a genome-wide chromatin immunoprecipitation (ChIP) approach, we characterized the patterns of global recruitment (and consequently, affected genes) of HBx or its mutant, deficient in binding to p65/RelA, of the NF-κB complex, in the presence or experimental depletion of a master coregulator MTA1. We found that the overall recruitment of HBx increases following an interruption in the involvement of MTA1 and/or p65/RelA, suggesting that the levels or activities of MTA1 and p65/RelA represent two major modifiers of HBx recruitment to the human genome. Special attention was placed on targets with recruitment to transcription start sites (TSS), suggesting a probable contribution of the trans-regulatory processes, in influencing the expression of the putative HBx target genes. Representative target genes with TSS recruitment were validated by PCR and DNA gel electrophoresis and with quantitative PCR for analysis of resulting altered expression in ChIP samples. This is the first high- throughput analysis of the genome-wide recruitment of HBx to characterize the involvement of a master chromatin modifier as well as a transcription factor, which act to alter the ability of HBx to trans-regulate genes that may be involved in the development of vii HBV-associated HCC. This analysis provides novel insight into the identity of targeted genes that are trans-regulated by this viral protein and elucidates how these factors may contribute to the functions of HBx, a major risk factor for the development of HCC. viii Table of Contents Dedication………………………………………………………………………………....iv Acknowledgements………………………………………………………………………...v Abstract……………………………………………………………………………..…….vii Table of Contents……………………………………………………………………...…..ix List of Figures…………………………………………………………………………….xii List of Tables ……………………………………………………………………….……xiv List of Abbreviations………………………………………………………………….…..xv Chapter 1: Introduction……………………………………………………………………..1 1.1 Hepatitis B virus and Hepatocellular Carcinoma………...….....……………….1 1.1.1 Viral Oncology and HBV…………………..……………..………………1 1.1.2 HBV and HBx…………….……………...……………………………….2 1.1.3 HBx and HCC…………………...….…………………………………….3 1.2 Host-viral Interactions Associated with HBx……...……………………….…..9 1.2.1 MTA1 and HBx…………………………..………………..……………..9 1.2.2 NF-κB, MTA1 and HBx………………..…….…………………………12 1.3 Specific Aims………………..……...…………………………………………18 Chapter 2: Materials and Methods………………………………………………………...19 2.1 Cell Lines…………….………..…………………………..…………………..19 2.2 Cloning………………………...………………………………….…………...19 2.3 Lentiviral Vectors…………………………...………………………………...20 2.4 siRNA Experiments………..…………………….……………………………20 ix 2.5 Western Blotting Analysis………………………………………………..……21 2.6 FACS Analysis and Fluorescent Microscopy…….…………………………….21 2.7 Chromatin Immunoprecipitation and Sequencing…………….……….………..22 2.8 Computational Analyses…………………..………………………………...….22 2.8.1 Avadis NGS………………………………………………………….22 2.8.2 UCSC Genome Browser…………...………………………………..23 2.8.3 Ingenuity Pathway Analysis (IPA)………..………………………...24 2.9 PCR, DNA Gel Electrophoresis, and qPCR…..………...…….………………...24 Chapter 3: Results…………………………………………………………………………26 3.1 Introduction and Experimental Model…………………….……………………26 3.1.1 Introduction…………………………..……………………………..26 3.1.2 Experimental Strategy…………………………………………….....27 3.2 Global Genome-wide Targeting of HBx and the Role of MTA1…………….....40 3.2.1 Characteristics of Binding………….……………………………….40 3.2.2 MTA1 and p65 are Central to Genome-wide Targeting by IPA Network Analysis…………………….……….……………………………46 3.2.3 Genome-wide Targeting of HBx to Chromosomes…………..……..56 3.2.4 Unfiltered Heaviest HBx Recruitment……………………………….61 3.3 MTA1 Status Alters the Ability of HBx to Target Categories of Genes……..……61 3.3.1 Molecular and Cellular Functions and Canonical Pathways of Targeted Genes……….…………...……………………………….……...61 3.3.2 HBx Recruitment to Top Nodal Points of Networks…………….....71 x 3.4 HBx
Recommended publications
  • ABSTRACT ANGSTADT, ANDREA Y. Evaluation of the Genomic
    ABSTRACT ANGSTADT, ANDREA Y. Evaluation of the Genomic Aberrations in Canine Osteosarcoma and Their Resemblance to the Human Counterpart. (Under the direction of Dr. Matthew Breen). In the last decade the domestic dog has emerged as an ideal biomedical model of complex genetic diseases such as cancers. Cancer in the dog occurs spontaneously and several studies have concluded that human and canine cancers have similar characteristics such as presentation of disease, rate of metastases, genetic dysregulation, and survival rates. Furthermore, in the genomic era the dog genome was found more homologous in sequence conservation to humans than mice, making it a valuable model organism for genetic study in addition to pathophysiological analysis. Osteosarcoma (OS), the most commonly diagnosed malignant bone tumor in humans and dogs, is one such cancer that would benefit from comparative genomic analysis. In humans, OS is a rare cancer diagnosed in fewer than 1,000 people per year in the USA, while in the domestic dog population the annual number of new cases is estimated to far exceed 10,000. This high rate of disease occurrence in dogs provides a unique opportunity to study the genomic imbalances in canine OS and their translational value to human OS as a means to identify important alterations involved in disease etiology. OS in humans is characterized by extremely complex karyotypes which contain both structural changes (translocations and/or rearrangements) and DNA copy number changes. Metaphase and array comparative genomic hybridization (aCGH) has assisted in uncovering the genetic imbalances that are associated with human OS phenotype. In dog OS, previous low-resolution (10-20Mb) aCGH analysis identified a wide range of recurrent copy number aberrations (CNAs), indicative of a similar level of genomic instability to human OS.
    [Show full text]
  • PRODUCT SPECIFICATION Anti-C12orf43
    Anti-C12orf43 Product Datasheet Polyclonal Antibody PRODUCT SPECIFICATION Product Name Anti-C12orf43 Product Number HPA046148 Gene Description chromosome 12 open reading frame 43 Clonality Polyclonal Isotype IgG Host Rabbit Antigen Sequence Recombinant Protein Epitope Signature Tag (PrEST) antigen sequence: AWGLEQRPHVAGKPRAGAANSQLSTSQPSLRHKVNEHEQDGNELQTTPEF RAHVAKKLGALLDSFITISEAAKEPAKAKVQKVALEDDGFRLFFTSVPGG REKEESPQPR Purification Method Affinity purified using the PrEST antigen as affinity ligand Verified Species Human Reactivity Recommended IHC (Immunohistochemistry) Applications - Antibody dilution: 1:50 - 1:200 - Retrieval method: HIER pH6 WB (Western Blot) - Working concentration: 0.04-0.4 µg/ml ICC-IF (Immunofluorescence) - Fixation/Permeabilization: PFA/Triton X-100 - Working concentration: 0.25-2 µg/ml Characterization Data Available at atlasantibodies.com/products/HPA046148 Buffer 40% glycerol and PBS (pH 7.2). 0.02% sodium azide is added as preservative. Concentration Lot dependent Storage Store at +4°C for short term storage. Long time storage is recommended at -20°C. Notes Gently mix before use. Optimal concentrations and conditions for each application should be determined by the user. For protocols, additional product information, such as images and references, see atlasantibodies.com. Product of Sweden. For research use only. Not intended for pharmaceutical development, diagnostic, therapeutic or any in vivo use. No products from Atlas Antibodies may be resold, modified for resale or used to manufacture commercial products without prior written approval from Atlas Antibodies AB. Warranty: The products supplied by Atlas Antibodies are warranted to meet stated product specifications and to conform to label descriptions when used and stored properly. Unless otherwise stated, this warranty is limited to one year from date of sales for products used, handled and stored according to Atlas Antibodies AB's instructions.
    [Show full text]
  • Purkinje Cell Migration Disorder By
    CEREBELLAR CORTICOGENESIS IN THE LYSOSOMAL ACID PHOSPHATASE (ACP2) MUTANT MICE: PURKINJE CELL MIGRATION DISORDER BY NILOUFAR ASHTARI A Thesis Submitted to the Faculty of Graduate Studies of The University of Manitoba in Partial Fulfilment of the Requirements for the Degree of MASTER OF SCIENCE Department of Human Anatomy and Cell Science University of Manitoba Winnipeg, Manitoba Copyright © 2017 by Niloufar Ashtari 1 Abstract In a mutant mouse called nax as the result of mutation in Lysosomal Acid phosphatase (Acp2), layers of the cerebellar cortex are impaired and monolayer Purkinje cells (Pcs) turn to multi-layered Pcs that ectopically invade the molecular layer. We investigated reelin-Dab1 signaling as an important pathway for Pcs migration and monolayer formation in cerebellum. ERK1/2 is a member of mitogen activated kinases family and suggested to be a downstream of reelin signaling. We hypothesize that the establishment of mono-layered Pcs rely on reelin through ERK1/2 pathway. Acp2 mutant mice were used for this study and molecular expression and distribution were assessed by immunohistochemistry, RT-PCR, western blotting, and cell culture. Results suggest that reelin may modulate the ERK1/2 expression, thus lower expression of reelin and higher phosphorylation of Dab1 leads to over expression of the ERK1/2 that causes the Pcs to over migrate and form multilayer in nax cerebellar cortex. i TABLE OF CONTENTS LISTOFABBREVIATIONS………………………………………………..…... IV LIST OF TABLES……………………………………...…………………...….. Vii LIST OF FIGURES…………………………………………………….………. Viii CHAPTER 1: INTRODUCTION…………………………………….………… 1 1.1 Cerebellum ……………………………………………………........………. 1 1.2 Development of Central Nervous System………………………………….. 2 1.3 Development of the cerebellum………………………………….................. 3 1.4 Specification of cerebellar germinal zones………………………………….
    [Show full text]
  • Inherited Neuropathies
    407 Inherited Neuropathies Vera Fridman, MD1 M. M. Reilly, MD, FRCP, FRCPI2 1 Department of Neurology, Neuromuscular Diagnostic Center, Address for correspondence Vera Fridman, MD, Neuromuscular Massachusetts General Hospital, Boston, Massachusetts Diagnostic Center, Massachusetts General Hospital, Boston, 2 MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology Massachusetts, 165 Cambridge St. Boston, MA 02114 and The National Hospital for Neurology and Neurosurgery, Queen (e-mail: [email protected]). Square, London, United Kingdom Semin Neurol 2015;35:407–423. Abstract Hereditary neuropathies (HNs) are among the most common inherited neurologic Keywords disorders and are diverse both clinically and genetically. Recent genetic advances have ► hereditary contributed to a rapid expansion of identifiable causes of HN and have broadened the neuropathy phenotypic spectrum associated with many of the causative mutations. The underlying ► Charcot-Marie-Tooth molecular pathways of disease have also been better delineated, leading to the promise disease for potential treatments. This chapter reviews the clinical and biological aspects of the ► hereditary sensory common causes of HN and addresses the challenges of approaching the diagnostic and motor workup of these conditions in a rapidly evolving genetic landscape. neuropathy ► hereditary sensory and autonomic neuropathy Hereditary neuropathies (HN) are among the most common Select forms of HN also involve cranial nerves and respiratory inherited neurologic diseases, with a prevalence of 1 in 2,500 function. Nevertheless, in the majority of patients with HN individuals.1,2 They encompass a clinically heterogeneous set there is no shortening of life expectancy. of disorders and vary greatly in severity, spanning a spectrum Historically, hereditary neuropathies have been classified from mildly symptomatic forms to those resulting in severe based on the primary site of nerve pathology (myelin vs.
    [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]
  • 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]
  • Download Download
    Supplementary Figure S1. Results of flow cytometry analysis, performed to estimate CD34 positivity, after immunomagnetic separation in two different experiments. As monoclonal antibody for labeling the sample, the fluorescein isothiocyanate (FITC)- conjugated mouse anti-human CD34 MoAb (Mylteni) was used. Briefly, cell samples were incubated in the presence of the indicated MoAbs, at the proper dilution, in PBS containing 5% FCS and 1% Fc receptor (FcR) blocking reagent (Miltenyi) for 30 min at 4 C. Cells were then washed twice, resuspended with PBS and analyzed by a Coulter Epics XL (Coulter Electronics Inc., Hialeah, FL, USA) flow cytometer. only use Non-commercial 1 Supplementary Table S1. Complete list of the datasets used in this study and their sources. GEO Total samples Geo selected GEO accession of used Platform Reference series in series samples samples GSM142565 GSM142566 GSM142567 GSM142568 GSE6146 HG-U133A 14 8 - GSM142569 GSM142571 GSM142572 GSM142574 GSM51391 GSM51392 GSE2666 HG-U133A 36 4 1 GSM51393 GSM51394 only GSM321583 GSE12803 HG-U133A 20 3 GSM321584 2 GSM321585 use Promyelocytes_1 Promyelocytes_2 Promyelocytes_3 Promyelocytes_4 HG-U133A 8 8 3 GSE64282 Promyelocytes_5 Promyelocytes_6 Promyelocytes_7 Promyelocytes_8 Non-commercial 2 Supplementary Table S2. Chromosomal regions up-regulated in CD34+ samples as identified by the LAP procedure with the two-class statistics coded in the PREDA R package and an FDR threshold of 0.5. Functional enrichment analysis has been performed using DAVID (http://david.abcc.ncifcrf.gov/)
    [Show full text]
  • Novel Insights Into the Thaumarchaeota in the Deepest Oceans: Their Metabolism and Potential Adaptation Mechanisms
    Zhong et al. Microbiome (2020) 8:78 https://doi.org/10.1186/s40168-020-00849-2 RESEARCH Open Access Novel insights into the Thaumarchaeota in the deepest oceans: their metabolism and potential adaptation mechanisms Haohui Zhong1,2, Laura Lehtovirta-Morley3, Jiwen Liu1,2, Yanfen Zheng1, Heyu Lin1, Delei Song1, Jonathan D. Todd3, Jiwei Tian4 and Xiao-Hua Zhang1,2,5* Abstract Background: Marine Group I (MGI) Thaumarchaeota, which play key roles in the global biogeochemical cycling of nitrogen and carbon (ammonia oxidizers), thrive in the aphotic deep sea with massive populations. Recent studies have revealed that MGI Thaumarchaeota were present in the deepest part of oceans—the hadal zone (depth > 6000 m, consisting almost entirely of trenches), with the predominant phylotype being distinct from that in the “shallower” deep sea. However, little is known about the metabolism and distribution of these ammonia oxidizers in the hadal water. Results: In this study, metagenomic data were obtained from 0–10,500 m deep seawater samples from the Mariana Trench. The distribution patterns of Thaumarchaeota derived from metagenomics and 16S rRNA gene sequencing were in line with that reported in previous studies: abundance of Thaumarchaeota peaked in bathypelagic zone (depth 1000–4000 m) and the predominant clade shifted in the hadal zone. Several metagenome-assembled thaumarchaeotal genomes were recovered, including a near-complete one representing the dominant hadal phylotype of MGI. Using comparative genomics, we predict that unexpected genes involved in bioenergetics, including two distinct ATP synthase genes (predicted to be coupled with H+ and Na+ respectively), and genes horizontally transferred from other extremophiles, such as those encoding putative di-myo-inositol-phosphate (DIP) synthases, might significantly contribute to the success of this hadal clade under the extreme condition.
    [Show full text]
  • (P -Value<0.05, Fold Change≥1.4), 4 Vs. 0 Gy Irradiation
    Table S1: Significant differentially expressed genes (P -Value<0.05, Fold Change≥1.4), 4 vs. 0 Gy irradiation Genbank Fold Change P -Value Gene Symbol Description Accession Q9F8M7_CARHY (Q9F8M7) DTDP-glucose 4,6-dehydratase (Fragment), partial (9%) 6.70 0.017399678 THC2699065 [THC2719287] 5.53 0.003379195 BC013657 BC013657 Homo sapiens cDNA clone IMAGE:4152983, partial cds. [BC013657] 5.10 0.024641735 THC2750781 Ciliary dynein heavy chain 5 (Axonemal beta dynein heavy chain 5) (HL1). 4.07 0.04353262 DNAH5 [Source:Uniprot/SWISSPROT;Acc:Q8TE73] [ENST00000382416] 3.81 0.002855909 NM_145263 SPATA18 Homo sapiens spermatogenesis associated 18 homolog (rat) (SPATA18), mRNA [NM_145263] AA418814 zw01a02.s1 Soares_NhHMPu_S1 Homo sapiens cDNA clone IMAGE:767978 3', 3.69 0.03203913 AA418814 AA418814 mRNA sequence [AA418814] AL356953 leucine-rich repeat-containing G protein-coupled receptor 6 {Homo sapiens} (exp=0; 3.63 0.0277936 THC2705989 wgp=1; cg=0), partial (4%) [THC2752981] AA484677 ne64a07.s1 NCI_CGAP_Alv1 Homo sapiens cDNA clone IMAGE:909012, mRNA 3.63 0.027098073 AA484677 AA484677 sequence [AA484677] oe06h09.s1 NCI_CGAP_Ov2 Homo sapiens cDNA clone IMAGE:1385153, mRNA sequence 3.48 0.04468495 AA837799 AA837799 [AA837799] Homo sapiens hypothetical protein LOC340109, mRNA (cDNA clone IMAGE:5578073), partial 3.27 0.031178378 BC039509 LOC643401 cds. [BC039509] Homo sapiens Fas (TNF receptor superfamily, member 6) (FAS), transcript variant 1, mRNA 3.24 0.022156298 NM_000043 FAS [NM_000043] 3.20 0.021043295 A_32_P125056 BF803942 CM2-CI0135-021100-477-g08 CI0135 Homo sapiens cDNA, mRNA sequence 3.04 0.043389246 BF803942 BF803942 [BF803942] 3.03 0.002430239 NM_015920 RPS27L Homo sapiens ribosomal protein S27-like (RPS27L), mRNA [NM_015920] Homo sapiens tumor necrosis factor receptor superfamily, member 10c, decoy without an 2.98 0.021202829 NM_003841 TNFRSF10C intracellular domain (TNFRSF10C), mRNA [NM_003841] 2.97 0.03243901 AB002384 C6orf32 Homo sapiens mRNA for KIAA0386 gene, partial cds.
    [Show full text]
  • Appendix 2. Significantly Differentially Regulated Genes in Term Compared with Second Trimester Amniotic Fluid Supernatant
    Appendix 2. Significantly Differentially Regulated Genes in Term Compared With Second Trimester Amniotic Fluid Supernatant Fold Change in term vs second trimester Amniotic Affymetrix Duplicate Fluid Probe ID probes Symbol Entrez Gene Name 1019.9 217059_at D MUC7 mucin 7, secreted 424.5 211735_x_at D SFTPC surfactant protein C 416.2 206835_at STATH statherin 363.4 214387_x_at D SFTPC surfactant protein C 295.5 205982_x_at D SFTPC surfactant protein C 288.7 1553454_at RPTN repetin solute carrier family 34 (sodium 251.3 204124_at SLC34A2 phosphate), member 2 238.9 206786_at HTN3 histatin 3 161.5 220191_at GKN1 gastrokine 1 152.7 223678_s_at D SFTPA2 surfactant protein A2 130.9 207430_s_at D MSMB microseminoprotein, beta- 99.0 214199_at SFTPD surfactant protein D major histocompatibility complex, class II, 96.5 210982_s_at D HLA-DRA DR alpha 96.5 221133_s_at D CLDN18 claudin 18 94.4 238222_at GKN2 gastrokine 2 93.7 1557961_s_at D LOC100127983 uncharacterized LOC100127983 93.1 229584_at LRRK2 leucine-rich repeat kinase 2 HOXD cluster antisense RNA 1 (non- 88.6 242042_s_at D HOXD-AS1 protein coding) 86.0 205569_at LAMP3 lysosomal-associated membrane protein 3 85.4 232698_at BPIFB2 BPI fold containing family B, member 2 84.4 205979_at SCGB2A1 secretoglobin, family 2A, member 1 84.3 230469_at RTKN2 rhotekin 2 82.2 204130_at HSD11B2 hydroxysteroid (11-beta) dehydrogenase 2 81.9 222242_s_at KLK5 kallikrein-related peptidase 5 77.0 237281_at AKAP14 A kinase (PRKA) anchor protein 14 76.7 1553602_at MUCL1 mucin-like 1 76.3 216359_at D MUC7 mucin 7,
    [Show full text]
  • Review Article Mouse Homologues of Human Hereditary Disease
    I Med Genet 1994;31:1-19 I Review article J Med Genet: first published as 10.1136/jmg.31.1.1 on 1 January 1994. Downloaded from Mouse homologues of human hereditary disease A G Searle, J H Edwards, J G Hall Abstract involve homologous loci. In this respect our Details are given of 214 loci known to be genetic knowledge of the laboratory mouse associated with human hereditary dis- outstrips that for all other non-human mam- ease, which have been mapped on both mals. The 829 loci recently assigned to both human and mouse chromosomes. Forty human and mouse chromosomes3 has now two of these have pathological variants in risen to 900, well above comparable figures for both species; in general the mouse vari- other laboratory or farm animals. In a previous ants are similar in their effects to the publication,4 102 loci were listed which were corresponding human ones, but excep- associated with specific human disease, had tions include the Dmd/DMD and Hprt/ mouse homologues, and had been located in HPRT mutations which cause little, if both species. The number has now more than any, harm in mice. Possible reasons for doubled (table 1A). Of particular interest are phenotypic differences are discussed. In those which have pathological variants in both most pathological variants the gene pro- the mouse and humans: these are listed in table duct seems to be absent or greatly 2. Many other pathological mutations have reduced in both species. The extensive been detected and located in the mouse; about data on conserved segments between half these appear to lie in conserved chromo- human and mouse chromosomes are somal segments.
    [Show full text]
  • Target Gene Gene Description Validation Diana Miranda
    Supplemental Table S1. Mmu-miR-183-5p in silico predicted targets. TARGET GENE GENE DESCRIPTION VALIDATION DIANA MIRANDA MIRBRIDGE PICTAR PITA RNA22 TARGETSCAN TOTAL_HIT AP3M1 adaptor-related protein complex 3, mu 1 subunit V V V V V V V 7 BTG1 B-cell translocation gene 1, anti-proliferative V V V V V V V 7 CLCN3 chloride channel, voltage-sensitive 3 V V V V V V V 7 CTDSPL CTD (carboxy-terminal domain, RNA polymerase II, polypeptide A) small phosphatase-like V V V V V V V 7 DUSP10 dual specificity phosphatase 10 V V V V V V V 7 MAP3K4 mitogen-activated protein kinase kinase kinase 4 V V V V V V V 7 PDCD4 programmed cell death 4 (neoplastic transformation inhibitor) V V V V V V V 7 PPP2R5C protein phosphatase 2, regulatory subunit B', gamma V V V V V V V 7 PTPN4 protein tyrosine phosphatase, non-receptor type 4 (megakaryocyte) V V V V V V V 7 EZR ezrin V V V V V V 6 FOXO1 forkhead box O1 V V V V V V 6 ANKRD13C ankyrin repeat domain 13C V V V V V V 6 ARHGAP6 Rho GTPase activating protein 6 V V V V V V 6 BACH2 BTB and CNC homology 1, basic leucine zipper transcription factor 2 V V V V V V 6 BNIP3L BCL2/adenovirus E1B 19kDa interacting protein 3-like V V V V V V 6 BRMS1L breast cancer metastasis-suppressor 1-like V V V V V V 6 CDK5R1 cyclin-dependent kinase 5, regulatory subunit 1 (p35) V V V V V V 6 CTDSP1 CTD (carboxy-terminal domain, RNA polymerase II, polypeptide A) small phosphatase 1 V V V V V V 6 DCX doublecortin V V V V V V 6 ENAH enabled homolog (Drosophila) V V V V V V 6 EPHA4 EPH receptor A4 V V V V V V 6 FOXP1 forkhead box P1 V
    [Show full text]