Supplementary Table 8. Genes Expressed in HNSCC and That Map to Regions Exhibiting Recurrent Genomic Amplification in Head and Neck Carcinomas

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Table 8. Genes Expressed in HNSCC and That Map to Regions Exhibiting Recurrent Genomic Amplification in Head and Neck Carcinomas Supplementary Table 8. Genes expressed in HNSCC and that map to regions exhibiting recurrent genomic amplification in head and neck carcinomas. Note that these ORESTES contigs do not contain sequences derived from non-tumour sequences. Legends to table “Tissue” corresponds to the origin of the sequenced EST: Thy= thyroid; La= larynx; Pha= pharynx; OC= oral cavity.From Locus Link (http://www.ncbi.nlm.nih.gov/LocusLink/): IMP=Inferred from mutant phenotype; IGI=Inferred from genetic interaction; IPI=Inferred from physical interaction; ISS=Inferred from sequence or structural similarity; IDA=Inferred from direct assay; IEP=Inferred from expression pattern; IEA=Inferred from electronic annotation; TAS=Traceable author statement; NAS=Non-traceable author statement; NR=Not recorded; E=Experimental evidence; P=Predicted/computed; GC: from Genecards (http://bioinformatics.weizmann.ac.il/cards/) Refseq description Cytogenetic Tissue Cluster Refseq accession Group/ Mapping Category absent in melanoma 2 (AIM2) 1q22 Oc R2_CLUSTER_12274 gi|4757733|ref|NM_004833| Cell communicatio n/ Response to external stimulus (GC) ACN9 homolog (S. cerevisiae) (ACN9) 7q22.1 Pha R2_CLUSTER_36704 gi|9910179|ref|NM_020186| Unknown function/ Unknown adrenergic, beta, receptor kinase 1 11q13 Oc R2_CLUSTER_21698 gi|6138971|ref|NM_001619| Cell (ADRBK1) communicatio n/ Signal transduction (IEA, TAS) AE(adipocyte enhancer)-binding protein 12p12.3 Oc, Thy R2_CLUSTER_42981 gi|34222204|ref|NM_153207| Unknown 2 (AEBP2) function/ Unknown aldehyde dehydrogenase 3 family, 11q13 Pha R2_CLUSTER_34245 gi|4580414|ref|NM_000695 | Cell member B2 (ALDH3B2) metabolism/ Lipid metabolism (TAS) aminoadipate-semialdehyde synthase 7q31.3 La R2_CLUSTER_36371 gi|13027639|ref|NM_005763| Cell (AASS) R2_CLUSTER_4381 gi|13027639|ref|NM_005763| Structure, Motility & Transport / Transport (IEA) ankylosis, progressive homolog (mouse) 5p15 Oc, La R2_CLUSTER_26599 gi|34452701|ref|NM_054027| Cell structure, (ANKH), transcript variant 2 R2_CLUSTER_20351 motility & transport/ Transport (IEA) ash1 (absent, small, or homeotic)-like 1q22 Oc R2_CLUSTER_34747 gi|8922080|ref|NM_018489| Cell (Drosophila) (ASH1L) metabolism/ Transcription regulation (TAS) atrophin-1 interacting protein 1 (AIP1) 7q21 Oc, La R2_CLUSTER_49969 gi|27436956|ref|NM_012301| Cell R2_CLUSTER_32294 communicatio n/ Signal transduction (GC) calcitonin receptor-like (CALCRL) 2q32 Oc, La R2_CLUSTER_29431 gi|34878691|ref|NM_005795| Cell R2_CLUSTER_45756 communicatio n/ Signal transduction (TAS, IEA) calcium channel, voltage-dependent, 7q21-q22 La, Thy R2_CLUSTER_32874 gi|4757893|ref|NM_000722| Cell structure, alpha 2/delta subunit 1 (CACNA2D1) R2_CLUSTER_35171 motility & transport/ Transport (IEA) calcium channel, voltage-dependent, L 12p13 Pha R2_CLUSTER_26662 gi|27597079|ref|NM_000719| Cell structure, type, alpha 1C subunit (CACNA1C) motility & transport/ Transport (E) calneuron 1 (CALN1) 7q11 Oc, La R2_CLUSTER_29667 gi|13899314|ref|NM_031468| Unknown R2_CLUSTER_27031 function / Unknown calsequestrin 1 (fast-twitch, skeletal 1q21 La R2_CLUSTER_23179 gi|21536273|ref|NM_001231| Cell structure, muscle) (CASQ1) motility & transport/ Motility (IEA) carnitine O-octanoyltransferase (CROT) 7q21 La, Oc R2_CLUSTER_44655 gi|31542324|ref|NM_021151| Cell R2_CLUSTER_36337 metabolism/ General metabolism (GC) Cas-Br-M (murine) ecotropic retroviral 7q22 Oc R2_CLUSTER_23343 gi|13376203|ref|NM_024814| Unknown transforming sequence-like 1 (CBLL1) function / Unknown CCR4-NOT transcription complex, 7q22-qter Oc R2_CLUSTER_13443 gi|7019466|ref|NM_013316| Unknown subunit 4 (CNOT4) function / Unknown CD84 antigen (leukocyte antigen) 1q24 Oc R2_CLUSTER_48640 gi|4502686|ref|NM_003874| Cell (CD84) communicatio n/ Response to external stimulus (GC) chloride channel 2 (CLCN2) 3q27-q28 Oc, Thy R2_CLUSTER_50739 gi|5803001|ref|NM_004366| Cell structure, motility & transport/ Transport (IEA) choline kinase (CHK) 11q13 La R2_CLUSTER_46668 gi|4557454|ref|NM_001277| Cell metabolism/ Lipid metabolism (TAS) cholinergic receptor, nicotinic, alpha 2q24-q32 Oc R2_CLUSTER_46191 gi|4557456|ref|NM_000079| Cell structure, polypeptide 1 (muscle) (CHRNA1) motility & transport/ Transport (IEA, TAS) chromosome 1 open reading frame 10 1q21 Pha, La R2_CLUSTER_6197 gi|7706634|ref|NM_016190| Unknown (C1orf10) function/ Unknown chromosome 11 open reading frame 23 11q13 La R2_CLUSTER_31954 gi|13489082|ref|NM_018312| Unknown (C11orf23) R2_CLUSTER_40608 function/ Unknown chromosome 12 open reading frame 5 12p13 Oc R2_CLUSTER_33339 gi|9966848|ref|NM_020375| Cell (C12orf5) metabolism (IEA) coatomer protein complex, subunit 7q32 Oc R2_CLUSTER_28160 gi|6912319|ref|NM_012133| Cell gamma 2 (COPG2) R2_CLUSTER_49430 Structure, Motility & Transport / Transport (IEA) cold shock domain protein A (CSDA) 12p13 La R2_CLUSTER_23473 gi|21359983|ref|NM_003651| Cell metabolism/ Transcription regulation (E) component of oligomeric golgi complex 7q22-q31 La R2_CLUSTER_24679 gi|32481215|ref|NM_006348| Cell structure, 5 (COG5), transcript variant 1 motility & transport/ Transport (IEA, NAS) contactin associated protein-like 2 7q35-q36 Pha R2_CLUSTER_32315 gi|21071040|ref|NM_014141| Cell (CNTNAP2) communicatio n / Cell adhesion (IEA) cut-like 1, CCAAT displacement protein 7q22 La R2_CLUSTER_34860 gi|31652239|ref|NM_181552| Cell (Drosophila) (CUTL1), transcript variant metabolism/ 1 Transcription regulation (E) cytochrome b reductase 1 (CYBRD1) 2q31 Oc R2_CLUSTER_20640 gi|19923602|ref|NM_024843| Cell structure, motility & transport/ Transport (IEA) cytoplasmic linker 2 (CYLN2), transcript 7q113 La, Thy R2_CLUSTER_34145 gi|14702160|ref|NM_003388| Unknown variant 1 R2_CLUSTER_45880 function / Unknown deltex homolog 2 (Drosophila) (DTX2) 7q113 Oc, La R2_CLUSTER_18029 gi|24308252|ref|NM_020892| Unknown R2_CLUSTER_29201 function / Unknown discs, large (Drosophila) homolog 1 3q29 Oc,La R2_CLUSTER_36831 gi|4758161|ref|NM_004087| Cell (DLG1) R2_CLUSTER_29359 communicatio n/ Signal transduction (TAS) distal-less homeo box 6 (DLX6) 7q22 Oc R2_CLUSTER_45223 gi|41147638|ref|XM_371934| Cell metabolism/ Transcription regulation (TAS, IEA) DnaJ (Hsp40) homolog, subfamily B, 7q36 Oc R2_CLUSTER_19204 gi|24234717|ref|NM_058246| Cell member 6 (DNAJB6), transcript variant 1 metabolism / Protein metabolism (NAS) E3 ubiquitin ligase SMURF1 (SMURF1), 7q21-q31 La, Oc R2_CLUSTER_32334 gi|31317291|ref|NM_020429| Cell transcript variant 1 R2_CLUSTER_33552 metabolism/ R2_CLUSTER_3643 Protein metabolism (IDA) elastin (supravalvular aortic stenosis, 7q113 La R2_CLUSTER_34507 gi|5881412|ref|NM_000501| Cell Williams-Beuren syndrome) (ELN) Structure, Motility & Transport / Intracellular matrix, membrane, cytoskeleton (IEA) ELKS protein (ELKS) 12p13 Oc R2_CLUSTER_9609 gi|38045899|ref|NM_015064| Unknown function/ Unknown ems1 sequence (mammary tumor and 11q13 La R2_CLUSTER_30031 gi|20357551|ref|NM_005231| Cell structure, squamous cell carcinoma-associated motility & (p80/85 src substrate) (EMS1), transcript transport/ variant 1 Extracellular matrix, membrane, cytoskeleton (GC) enhancer of zeste homolog 2 7q35-q36 La R2_CLUSTER_18289 gi|23510382|ref|NM_004456| Cell (Drosophila) (EZH2), transcript variant 1 Metabolism / Transcription regulation (GC) ets variant gene 5 (ets-related molecule) 3q28 Oc, Thy R2_CLUSTER_40729 gi|4758315|ref|NM_004454| Cell (ETV5) metabolism/ Transcription regulation (IEA, TAS) ets variant gene 6 (TEL oncogene) 12p13 La R2_CLUSTER_38631 gi|41872473|ref|NM_001987| Cell (ETV6) R2_CLUSTER_39962 metabolism/ Transcription regulation (TAS, IEA) FK506 binding protein 2, 13kDa 11q13-q13 La R2_CLUSTER_28402 gi|17149841|ref|NM_004470| Cell (FKBP2), transcript variant 1 metabolism/ Protein metabolism (IEA) glutaminase (GLS) 2q32-q34 Pha R2_CLUSTER_51296 gi|21361451|ref|NM_014905| Cell metabolism/ General metabolism (IEA, NAS) guanine nucleotide binding protein (G 7q21 La R2_CLUSTER_31558 gi|33946323|ref|NM_002069| Cell protein), alpha inhibiting activity communicatio polypeptide 1(GNAI1) n/ Signal transduction (IEA) guanine nucleotide binding protein (G 3q27 La R2_CLUSTER_28836 gi|20357531|ref|NM_021629| Cell protein), beta polypeptide 4 (GNB4) communicatio n/ Signal transduction (IEA) H2A histone family, member J (H2AFJ), 12p12 La R2_CLUSTER_35496 gi|29553976|ref|NM_018267| Cell structure, transcript variant 1 motility & transport/ Intracellular structure & organization (GC) HRD1 protein (HRD1), transcript variant 11q13 La R2_CLUSTER_30132 gi|27436926|ref|NM_172230| Unknown 2 function/ Unknown hypothetical 55 kDa protein F09G8 in 11q13 Thy, Oc R2_CLUSTER_19759 gi|21687174|ref|NM_145309| Unknown chromosome III (LOC220074) R2_CLUSTER_37146 function/ Unknown hypothetical protein DKFZp434O0515 2q31 La R2_CLUSTER_37514 gi|31342410|ref|NM_178123| Unknown (DKFZp434O0515) function/ Unknown hypothetical protein DKFZp761P1010 12p131 Oc R2_CLUSTER_12857 gi|8922178|ref|NM_018423| Cell (DKFZp761P1010) communicatio n/ Signal transduction (GC) hypothetical protein FLJ10204 8q243 La R2_CLUSTER_14014 gi|8922280|ref|NM_018024| Unknown (FLJ10204) function/ Unknown hypothetical protein FLJ10300 7q36 Oc, La R2_CLUSTER_35791 gi|21361686|ref|NM_018051| Unknown (FLJ10300) R2_CLUSTER_44172 function / Unknown hypothetical protein FLJ11280 1q21 Oc, La R2_CLUSTER_19625 gi|31377840|ref|NM_018379| Cell (FLJ11280) metabolism/ Transcription regulation (IEA) hypothetical protein FLJ12519 2q32 Oc R2_CLUSTER_47101 gi|29789282|ref|NM_032168| Cell structure, (FLJ12519) motility & transport/ Transport (IEA) hypothetical protein FLJ13195 similar to 7p11-q11 La R2_CLUSTER_25743
Recommended publications
  • Multi-Ancestry Genome-Wide Gene-Sleep Interactions Identify Novel
    bioRxiv preprint doi: https://doi.org/10.1101/2020.05.29.123505; this version posted May 31, 2020. 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. Multi-ancestry genome-wide gene-sleep interactions identify novel loci for blood pressure Heming Wang1,2,*, Raymond Noordam3,* , Brian E Cade1,2,*, Karen Schwander4,*, Thomas W Winkler5,*, Jiwon Lee1,*, Yun Ju Sung4,*, Amy R. Bentley6,*, Alisa K Manning2,7, Hugues Aschard8,9, Tuomas O Kilpeläinen10,11, Marjan Ilkov12, Michael R Brown13, Andrea R Horimoto14, Melissa Richard15, Traci M Bartz16, Dina Vojinovic17,18, Elise Lim19, Jovia L Nierenberg20, Yongmei Liu21, Kumaraswamynaidu Chitrala22, Tuomo Rankinen23, Solomon K Musani24, Nora Franceschini25, Rainer Rauramaa26, Maris Alver27,28, Phyllis Zee29, Sarah E Harris30, Peter J van der Most31, Ilja M Nolte31, Patricia B Munroe32,33, Nicholette D Palmer34, Brigitte Kühnel35,36, Stefan Weiss37,38, Wanqing Wen39, Kelly A Hall40, Leo-Pekka Lyytikäinen41,42, Jeff O'Connell43,44, Gudny Eiriksdottir12, Lenore J Launer22, Paul S de Vries13, Dan E Arking45, Han Chen13,46, Eric Boerwinkle13,47, Jose E Krieger14, Pamela J Schreiner48, Stephen S Sidney49, James M Shikany50, Kenneth Rice51, Yii-Der Ida Chen52, Sina A Gharib53, Joshua C Bis54, Annemarie I Luik17, M Arfan Ikram17,55, André G Uitterlinden17, Najaf Amin17, Hanfei Xu19, Daniel Levy19,56, Jiang He20, Kurt
    [Show full text]
  • Analysis of Trans Esnps Infers Regulatory Network Architecture
    Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2014 Anat Kreimer All rights reserved ABSTRACT Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer eSNPs are genetic variants associated with transcript expression levels. The characteristics of such variants highlight their importance and present a unique opportunity for studying gene regulation. eSNPs affect most genes and their cell type specificity can shed light on different processes that are activated in each cell. They can identify functional variants by connecting SNPs that are implicated in disease to a molecular mechanism. Examining eSNPs that are associated with distal genes can provide insights regarding the inference of regulatory networks but also presents challenges due to the high statistical burden of multiple testing. Such association studies allow: simultaneous investigation of many gene expression phenotypes without assuming any prior knowledge and identification of unknown regulators of gene expression while uncovering directionality. This thesis will focus on such distal eSNPs to map regulatory interactions between different loci and expose the architecture of the regulatory network defined by such interactions. We develop novel computational approaches and apply them to genetics-genomics data in human. We go beyond pairwise interactions to define network motifs, including regulatory modules and bi-fan structures, showing them to be prevalent in real data and exposing distinct attributes of such arrangements. We project eSNP associations onto a protein-protein interaction network to expose topological properties of eSNPs and their targets and highlight different modes of distal regulation.
    [Show full text]
  • Cisplatin and Phenanthriplatin Modulate Long-Noncoding
    www.nature.com/scientificreports OPEN Cisplatin and phenanthriplatin modulate long‑noncoding RNA expression in A549 and IMR90 cells revealing regulation of microRNAs, Wnt/β‑catenin and TGF‑β signaling Jerry D. Monroe1,2, Satya A. Moolani2,3, Elvin N. Irihamye2,4, Katheryn E. Lett1, Michael D. Hebert1, Yann Gibert1* & Michael E. Smith2* The monofunctional platinum(II) complex, phenanthriplatin, acts by blocking transcription, but its regulatory efects on long‑noncoding RNAs (lncRNAs) have not been elucidated relative to traditional platinum‑based chemotherapeutics, e.g., cisplatin. Here, we treated A549 non‑small cell lung cancer and IMR90 lung fbroblast cells for 24 h with either cisplatin, phenanthriplatin or a solvent control, and then performed microarray analysis to identify regulated lncRNAs. RNA22 v2 microRNA software was subsequently used to identify microRNAs (miRNAs) that might be suppressed by the most regulated lncRNAs. We found that miR‑25‑5p, ‑30a‑3p, ‑138‑5p, ‑149‑3p, ‑185‑5p, ‑378j, ‑608, ‑650, ‑708‑5p, ‑1253, ‑1254, ‑4458, and ‑4516, were predicted to target the cisplatin upregulated lncRNAs, IMMP2L‑1, CBR3‑1 and ATAD2B‑5, and the phenanthriplatin downregulated lncRNAs, AGO2‑1, COX7A1‑2 and SLC26A3‑1. Then, we used qRT‑PCR to measure the expression of miR‑25‑5p, ‑378j, ‑4516 (A549) and miR‑149‑3p, ‑608, and ‑4458 (IMR90) to identify distinct signaling efects associated with cisplatin and phenanthriplatin. The signaling pathways associated with these miRNAs suggests that phenanthriplatin may modulate Wnt/β‑catenin and TGF‑β signaling through the MAPK/ ERK and PTEN/AKT pathways diferently than cisplatin. Further, as some of these miRNAs may be subject to dissimilar lncRNA targeting in A549 and IMR90 cells, the monofunctional complex may not cause toxicity in normal lung compared to cancer cells by acting through distinct lncRNA and miRNA networks.
    [Show full text]
  • ASH1L Methyltransferase
    ASH1L (Ash1-Like Protein) CATALOG NO.: HMT-11-131 LOT NO.: DESCRIPTION: Human recombinant ASH1L (residues 2022-2302; Genbank Accession # NM_018489; MW = 59.5 kDa) expressed in E. coli with an N-terminal GST-tag. Catalyzes the transfer of methyl groups from S-adenosyl-L-methionine (SAM) to the ε-amino function of protein L-lysine residues, specifically lysine-36 of histone H3 (H3K36)1,2, a mark associated with active transcription. Activity has also been reported at H3K43. A large , multi-domain protein associated with actively transcribed regions of chromatin, ASH1L is the human homolog of Drosophila Ash1, a Trithorax group protein. Like its counterpart in Drosophila4, ASH1L contains a SET histone methyltransferase domain and has been found to play a role in the regulation of Hox gene expression3,5. Although the ASH1L SET domain has been shown in vitro to methylate histone peptides on lysine-4 of histone H3 (H3K4)3, in vivo or in vitro with nucleosomes as substrate, ASH1L is an H3K36 methyltransferase1,2,6. Recruitment of ASH1L by the ncRNA DBE-T to the chromosome 4q35 locus associated with FSHD (facioscapulohumeral muscular dystrophy) leads to increased H3K36me2 and inappropriate gene derepression at the FSHD locus6. In a possible positive feedback loop, ASH1L increases expression of DBE-T itself6, suggesting ASH1L’s methyltransferase activity or its interaction with DBE-T as potential therapeutic targets for FSHD. RBC’s ASH1L comprises the catalytic domain (AWS/SET/Post-SET) fused to GST. PURITY: >80% by SDS-PAGE ASSAY CONDITIONS: RBC’s ASH1L displays histone methyltransferase activity at concentrations of 62.5 nM-1 µM, 60 min.
    [Show full text]
  • Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin
    cells Review Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin Agnieszka Bochy ´nska,Juliane Lüscher-Firzlaff and Bernhard Lüscher * ID Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany; [email protected] (A.B.); jluescher-fi[email protected] (J.L.-F.) * Correspondence: [email protected]; Tel.: +49-241-8088850; Fax: +49-241-8082427 Received: 18 January 2018; Accepted: 27 February 2018; Published: 2 March 2018 Abstract: Regulation of gene expression is achieved by sequence-specific transcriptional regulators, which convey the information that is contained in the sequence of DNA into RNA polymerase activity. This is achieved by the recruitment of transcriptional co-factors. One of the consequences of co-factor recruitment is the control of specific properties of nucleosomes, the basic units of chromatin, and their protein components, the core histones. The main principles are to regulate the position and the characteristics of nucleosomes. The latter includes modulating the composition of core histones and their variants that are integrated into nucleosomes, and the post-translational modification of these histones referred to as histone marks. One of these marks is the methylation of lysine 4 of the core histone H3 (H3K4). While mono-methylation of H3K4 (H3K4me1) is located preferentially at active enhancers, tri-methylation (H3K4me3) is a mark found at open and potentially active promoters. Thus, H3K4 methylation is typically associated with gene transcription. The class 2 lysine methyltransferases (KMTs) are the main enzymes that methylate H3K4. KMT2 enzymes function in complexes that contain a necessary core complex composed of WDR5, RBBP5, ASH2L, and DPY30, the so-called WRAD complex.
    [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]
  • Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights Into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle
    animals Article Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle Masoumeh Naserkheil 1 , Abolfazl Bahrami 1 , Deukhwan Lee 2,* and Hossein Mehrban 3 1 Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj 77871-31587, Iran; [email protected] (M.N.); [email protected] (A.B.) 2 Department of Animal Life and Environment Sciences, Hankyong National University, Jungang-ro 327, Anseong-si, Gyeonggi-do 17579, Korea 3 Department of Animal Science, Shahrekord University, Shahrekord 88186-34141, Iran; [email protected] * Correspondence: [email protected]; Tel.: +82-31-670-5091 Received: 25 August 2020; Accepted: 6 October 2020; Published: 9 October 2020 Simple Summary: Hanwoo is an indigenous cattle breed in Korea and popular for meat production owing to its rapid growth and high-quality meat. Its yearling weight and carcass traits (backfat thickness, carcass weight, eye muscle area, and marbling score) are economically important for the selection of young and proven bulls. In recent decades, the advent of high throughput genotyping technologies has made it possible to perform genome-wide association studies (GWAS) for the detection of genomic regions associated with traits of economic interest in different species. In this study, we conducted a weighted single-step genome-wide association study which combines all genotypes, phenotypes and pedigree data in one step (ssGBLUP). It allows for the use of all SNPs simultaneously along with all phenotypes from genotyped and ungenotyped animals. Our results revealed 33 relevant genomic regions related to the traits of interest.
    [Show full text]
  • Congenital Disorders of Glycosylation from a Neurological Perspective
    brain sciences Review Congenital Disorders of Glycosylation from a Neurological Perspective Justyna Paprocka 1,* , Aleksandra Jezela-Stanek 2 , Anna Tylki-Szyma´nska 3 and Stephanie Grunewald 4 1 Department of Pediatric Neurology, Faculty of Medical Science in Katowice, Medical University of Silesia, 40-752 Katowice, Poland 2 Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland; [email protected] 3 Department of Pediatrics, Nutrition and Metabolic Diseases, The Children’s Memorial Health Institute, W 04-730 Warsaw, Poland; [email protected] 4 NIHR Biomedical Research Center (BRC), Metabolic Unit, Great Ormond Street Hospital and Institute of Child Health, University College London, London SE1 9RT, UK; [email protected] * Correspondence: [email protected]; Tel.: +48-606-415-888 Abstract: Most plasma proteins, cell membrane proteins and other proteins are glycoproteins with sugar chains attached to the polypeptide-glycans. Glycosylation is the main element of the post- translational transformation of most human proteins. Since glycosylation processes are necessary for many different biological processes, patients present a diverse spectrum of phenotypes and severity of symptoms. The most frequently observed neurological symptoms in congenital disorders of glycosylation (CDG) are: epilepsy, intellectual disability, myopathies, neuropathies and stroke-like episodes. Epilepsy is seen in many CDG subtypes and particularly present in the case of mutations
    [Show full text]
  • ING3 Is Essential for Asymmetric Cell Division During Mouse Oocyte Maturation
    ING3 Is Essential for Asymmetric Cell Division during Mouse Oocyte Maturation Shinnosuke Suzuki1, Yusuke Nozawa1, Satoshi Tsukamoto2, Takehito Kaneko3, Hiroshi Imai1, Naojiro Minami1* 1 Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan, 2 Laboratory Animal and Genome Sciences Section, National Institute of Radiological Sciences, Chiba, Japan, 3 Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan Abstract ING3 (inhibitor of growth family, member 3) is a subunit of the nucleosome acetyltransferase of histone 4 (NuA4) complex, which activates gene expression. ING3, which contains a plant homeodomain (PHD) motif that can bind to trimethylated lysine 4 on histone H3 (H3K4me3), is ubiquitously expressed in mammalian tissues and governs transcriptional regulation, cell cycle control, and apoptosis via p53-mediated transcription or the Fas/caspase-8 pathway. Thus, ING3 plays a number of important roles in various somatic cells. However, the role(s) of ING3 in germ cells remains unknown. Here, we show that loss of ING3 function led to the failure of asymmetric cell division and cortical reorganization in the mouse oocyte. Immunostaining showed that in fully grown germinal vesicle (GV) oocytes, ING3 localized predominantly in the GV. After germinal vesicle breakdown (GVBD), ING3 homogeneously localized in the cytoplasm. In oocytes where Ing3 was targeted by siRNA microinjection, we observed symmetric cell division during mouse oocyte maturation. In those oocytes, oocyte polarization was not established due to the failure to form an actin cap or a cortical granule-free domain (CGFD), the lack of which inhibited spindle migration. These features were among the main causes of abnormal symmetric cell division.
    [Show full text]
  • Molecular Effects of Isoflavone Supplementation Human Intervention Studies and Quantitative Models for Risk Assessment
    Molecular effects of isoflavone supplementation Human intervention studies and quantitative models for risk assessment Vera van der Velpen Thesis committee Promotors Prof. Dr Pieter van ‘t Veer Professor of Nutritional Epidemiology Wageningen University Prof. Dr Evert G. Schouten Emeritus Professor of Epidemiology and Prevention Wageningen University Co-promotors Dr Anouk Geelen Assistant professor, Division of Human Nutrition Wageningen University Dr Lydia A. Afman Assistant professor, Division of Human Nutrition Wageningen University Other members Prof. Dr Jaap Keijer, Wageningen University Dr Hubert P.J.M. Noteborn, Netherlands Food en Consumer Product Safety Authority Prof. Dr Yvonne T. van der Schouw, UMC Utrecht Dr Wendy L. Hall, King’s College London This research was conducted under the auspices of the Graduate School VLAG (Advanced studies in Food Technology, Agrobiotechnology, Nutrition and Health Sciences). Molecular effects of isoflavone supplementation Human intervention studies and quantitative models for risk assessment Vera van der Velpen Thesis submitted in fulfilment of the requirements for the degree of doctor at Wageningen University by the authority of the Rector Magnificus Prof. Dr M.J. Kropff, in the presence of the Thesis Committee appointed by the Academic Board to be defended in public on Friday 20 June 2014 at 13.30 p.m. in the Aula. Vera van der Velpen Molecular effects of isoflavone supplementation: Human intervention studies and quantitative models for risk assessment 154 pages PhD thesis, Wageningen University, Wageningen, NL (2014) With references, with summaries in Dutch and English ISBN: 978-94-6173-952-0 ABSTRact Background: Risk assessment can potentially be improved by closely linked experiments in the disciplines of epidemiology and toxicology.
    [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]
  • CNOT4 Antibody (Pab)
    21.10.2014CNOT4 antibody (pAb) Rabbit Anti-Human/Mouse/Rat CCR4-NOT Transcription Complex Subunit 4 (NOT4, NOT4H) Instruction Manual Catalog Number PK-AB718-4813 Synonyms CNOT4 Antibody: CCR4-NOT transcription complex subunit 4, NOT4, NOT4H Description CNOT4 is a component of the CCR4-NOT transcription complex, a complex that is implicated in the repression of RNA polymerase II transcription. In the CCR4-NOT complex, CNOT4 acts as an E3 ubiquitin-protein ligase and interacts with a subset of E2 ubiquitin-conjugating enzymes through a unique C4C4 RING domain. This E3 ligase activity was shown to be dependent on the selective and specific interaction with the ubiquitin conjugating enzyme UbcH5B. In yeast, mutations in CNOT4 that prevented its interaction with the UbcH5B homolog UBC4 caused increased sensitivity to hydroxyurea, heat shock, and hygromycin B, suggesting that CNOT4 and UbcH5B are involved in stress response in vivo. Multiple isoforms of CNOT4 are known to exist. Quantity 100 µg Source / Host Rabbit Immunogen CNOT4 antibody was raised against a 19 amino acid peptide near the amino terminus of the human CNOT4. Purification Method Affinity chromatography purified via peptide column. Clone / IgG Subtype Polyclonal antibody Species Reactivity Human, Mouse, Rat Specificity Formulation Antibody is supplied in PBS containing 0.02% sodium azide. Reconstitution During shipment, small volumes of antibody will occasionally become entrapped in the seal of the product vial. For products with volumes of 200 μl or less, we recommend gently tapping the vial on a hard surface or briefly centrifuging the vial in a tabletop centrifuge to dislodge any liquid in the container’s cap.
    [Show full text]