Yeast Genes Illuminate Human Cancer Gene Functions

Total Page:16

File Type:pdf, Size:1020Kb

Yeast Genes Illuminate Human Cancer Gene Functions Oncogene (2007) 26, 5373–5384 & 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00 www.nature.com/onc REVIEW MYST opportunities for growth control: yeast genes illuminate human cancer gene functions A Lafon, CS Chang, EM Scott, SJ Jacobson and L Pillus Section of Molecular Biology, Division of Biological Sciences, UCSD Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA The MYST family of histone acetyltransferases (HATs) genes were identified with potential roles in chromatin- was initially defined by human genes with disease mediated gene control, even modest degrees of similarity connections and by yeast genes identified for their role to the acetyl-CoA-binding regions were viewed with in epigenetic transcriptional silencing. Since then, many special interest as candidate HATs. new MYST genes have been discovered through genetic Such interest was particularly the case when mutants and genomic approaches. Characterization of the com- of SAS2 (something about silencing) were discovered as plexes through which MYST proteins act, regions of enhancers of mutations in the epigenetic transcriptional the genome to which they are targeted and biological silencer factor Sir1 (Reifsnyder et al., 1996) and consequences when they are disrupted, all deepen the suppressors of defects in cis-regulatory sequences for connections of MYST proteins to development, growth silent chromatin (Reifsnyder et al., 1996; Ehrenhofer- control and human cancers. Many of the insights into Murray et al., 1997). The observation that SAS2 and the MYST family function have come from studies in model closely related SAS3 yeast genes shared similarity to organisms. Herein, we review functions of two of the acetyl-CoA-binding domains, and even more significant founding MYST genes, yeast SAS2 and SAS3, and the similarity to two human genes, MOZ (Borrow et al., essential yeast MYST ESA1. Analysis of these genes in 1996) and TIP60 (Kamine et al., 1996), quickly led to yeast has defined roles for MYST proteins in transcrip- the recognition that together these genes defined a tional activation and silencing, and chromatin-mediated broadly conserved family that was named MYST for its boundary formation. They have further roles in DNA founding members (MOZ-YBF2/SAS3-SAS2-TIP60) damage repair and nuclear integrity. The observation that (Borrow et al., 1996; Reifsnyder et al., 1996). ESA1 MYST protein complexes share subunits with other HATs, was later identified as the third yeast MYST gene (Smith histone deacetylases and other key nuclear proteins, et al., 1998a; Clarke et al., 1999) with diverse functions: many with connections to human cancers, strengthens the it is essential for viability, and plays roles in transcrip- idea that coordinating distinct chromatin modifications tional activation, silencing, cell cycle progression and is critical for regulation. DNA damage repair. Oncogene (2007) 26, 5373–5384; doi:10.1038/sj.onc.1210606 The MYST domain – defined as the most conserved region in MYST family proteins – comprises a region Keywords: chromatin; transcription; Saccharomyces that includes both the sequences surrounding the acetyl- cerevisiae; silencing CoA-binding domain and in most enzymes an atypical C2HC zinc finger. Whereas the sequence of the MYST core domain is well conserved in Sas3 and Sas2, Esa1 lacks three of the four amino acids for zinc binding. Overview However, the crystal structure of Esa1 revealed that this region forms a classical TFIIIA-type zinc finger fold in Among the many chromatin modifications known, the absence of zinc suggesting a structurally conserved histone acetylation received particularly prominent early core domain with other MYST HATs (Yan et al., 2000). attention with the molecular identification of HAT1 and Besides the MYST domain, Sas3 and Esa1 contain GCN5 as genes encoding histone acetyltransferases additional conserved regions that are only found in (HATs) (Kleff et al., 1995; Brownell et al., 1996; subsets of the MYST family proteins, described below. Parthun et al., 1996). Although these two HATs have The early link between human MOZ, a recurrent different substrate specificities, initial sequence compar- translocation partner in several acute leukemias, and the isons revealed significant yet restricted similarity in three yeast genes implicated in chromatin-mediated regions predicted to bind acetyl-CoA. When additional transcriptional silencing raised the possibility that fundamentally conserved mechanisms might inform basic understanding between silencing mechanisms and Correspondence: Dr L Pillus, Section of Molecular Biology, Division of Biological Sciences, UCSD Moores Cancer Center, University of human disease. As further genomic and genetic analyses California, San Diego, La Jolla, CA 92093-0347, USA. proceeded, it became clear that the MYST family was E-mail: [email protected] broadly conserved. As of early 2007, several hundred Yeast MYST HATs and human cancer A Lafon et al 5374 related genomic sequences sharing significant sequence Here, their structural and functional distinctions are similarity over an B150 amino acid domain encompass- briefly highlighted in three categories: (1) definitions ing the acetyl-CoA-binding site have been annotated as of histone substrate specificity À of particular interest MYST genes (http://www.ncbi.nlm.nih.gov/entrez/query. are disease-relevant non-histone substrates such as fcgi?db ¼ Protein). Among MYST proteins of particular p53 in humans (Sykes et al., 2006; Tang et al., 2006), note are the Drosophila MOF gene that is essential only in (2) the MYST multi-protein complex(es) that modulate males for dosage compensation (Hilfiker et al., 1997) and enzymatic activity and promote coregulation with other the human MOF, MORF, MOZ and HBO1 genes that chromatin remodeling complexes through shared sub- have been implicated in multiple aspects of human units and (3) the gene-specific versus global acetylation development and disease (Avvakumov and Coˆ te´ ,2007; patterns that are of critical importance for disease and Rea et al., 2007). assessing potential success and side effect of therapeutic Many insights into MYST gene functions came initi- agents. These aspects are considered in more detail in ally from studies in model organisms, including Droso- other recent reviews (Utley and Coˆ te´ , 2003; Yang, 2004; phila, Caenorhabditis elegans, mouse, and the yeasts Altaf et al., 2007). We go on to address yeast MYSTs in Saccharomyces pombe and Saccharomyces cerevisiae the context of human disease, illustrated by mutant (described elsewhere in this issue and reviewed in Yang, phenotypes and conserved complex subunits. Further, 2004). This progress has come from the open-ended we consider what has been learned from genomic and nature of genetic screens, targeted gene deletion tech- proteomic surveys in yeast and speculate on what may niques and biochemical approaches combined with be learned from future studies that will be relevant for complete genome sequences and resources. understanding human health and disease. S. cerevisiae is a particularly rapid and informative genetic system for dissecting human protein function. For example, two-hybrid screening identified an inter- SAS3: critical functional links to GCN5 and H3 action of the MYST protein HBO1 with progesterone modifications receptor (PR) (Georgiakaki et al., 2006), a member of the family of steroid receptors. This observation SAS3 is arguably the least well-understood MYST gene prompted further analysis in human cells, which showed in yeast. In large part, this is because of the lack of that HBO1 enhancement of PR-dependent transcription extreme phenotypes observed upon deletion; indeed, required steroid receptor coactivator SRC-1 (Georgia- SAS3 was originally reported to have only minor kaki et al., 2006). Steroid receptors and their coactiva- functions in silencing at mating-type loci (Reifsnyder tors are key regulators of gene expression (Lonard and et al., 1996). O’Malley, 2006) and participate in signaling pathways Sas3 contains extensive stretches of acidic aspartate susceptible to misregulation in cancer cells. Yeast assays and glutamate residues in its C-terminus region, also can be designed also to study mechanisms of coactivator found in the human MYST HATs MOZ and MORF. function. For example, phenotypes conferred upon Although the functional significance of this domain in expression of hSRC-3 (Brown et al., 2003), GRIP1 the human proteins remains unclear, the acidic domain and SRC-1 (Anafi et al., 2000) revealed a role for Gcn5 of Sas3 is required for its physical interaction with the HAT complex components. These and other ingenious essential transcriptional elongation factor Spt16 through yeast assays will lend further insight into mechanisms two-hybrid screening (John et al., 2000). This suggests a underlying human MYST HAT function. The clinical function of Sas3 in transcriptional regulation mediated implications of these observations can be further tested by RNA polymerase II. in yeast screens to identify chemicals that interact with Although much remains to be learned of the biological steroid hormones and coactivators to modulate their processes regulated by SAS3, the determinants of its transcriptional output (Nishikawa et al., 1999) and molecular function have been well characterized, correlate mutations in key cancer-associated proteins including the multi-protein complex containing Sas3 with mutations that are clinically relevant, as was and the histone residues it targets. Sas3
Recommended publications
  • Expression Pattern and Level of ING5 Protein in Normal and Cancer Tissues
    ONCOLOGY LETTERS 17: 63-68, 2019 Expression pattern and level of ING5 protein in normal and cancer tissues XUE-FENG YANG1, DAO-FU SHEN1, SHUANG ZHAO1, TIAN-REN REN2, YANG GAO1, SHUAI SHI1, JI-CHENG WU1, HONG-ZHI SUN1 and HUA-CHUAN ZHENG1,3 1Cancer Center and Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001;2 Jilin Province Forestry Bureau, Linjiang, Jilin 134600; 3Institute of Life Sciences, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China Received February 21, 2016; Accepted February 13, 2017 DOI: 10.3892/ol.2018.9581 Abstract. Inhibitor of growth family 5 (ING5) functions as may be involved in cell regeneration and be associated with a type-II tumor suppressor gene and exerts an important role colorectal carcinogenesis. in DNA repair, apoptotic induction, proliferative inhibition, chromatin remodeling and the invasion process. In the present Introduction study, immunohistochemistry was performed to characterize the expression profile of ING5 protein on a tissue microarray Inhibitor of growth family 5 (ING5) is a member of the ING containing mouse and human normal tissues, and human protein family and functions as a type-II tumor suppressor cancer tissues, including hepatocellular (n=62), renal clear cell gene. Human ING5 is mapped to chromosome 2q37.3 and (n=62), pancreatic (n=62), esophageal squamous cell (n=45), encodes a 28-kDa protein with 240 amino acids (1). As indi- cervical squamous cell (n=31), breast (n=144), gastric (n=196), cated in Fig. 1, ING5 protein consists of a number of domains, colorectal (n=96), endometrial (n=96) and lung carcinoma including leucine zipper like (LZL), novel conserved region (n=192).
    [Show full text]
  • Expression of HAT1 and HDAC1, 2, 3 in Diffuse Large B-Cell Lymphomas, Peripheral T-Cell Lymphomas, and NK/T-Cell Lymphomas
    The Korean Journal of Pathology 2012; 46: 142-150 ▒ ORIGINAL ARTICLE ▒ http://dx.doi.org/10.4132/KoreanJPathol.2012.46.2.142 Expression of HAT1 and HDAC1, 2, 3 in Diffuse Large B-Cell Lymphomas, Peripheral T-Cell Lymphomas, and NK/T-Cell Lymphomas Soo Kee Min · Young Ho Koh1 Background: It has generally been proven that histone acetylation and deacetylation are involved Yunwoong Park1 · Hyo Jung Kim2 in the malignant transformation. To date, however, this has rarely been studied in cases of malig- Jinwon Seo · Hye-Rim Park nant lymphoma. Methods: We studied nine cases of reactive lymphoid hyperplasia, 78 cases of diffuse large B-cell lymphoma (DLBCL), 13 cases of peripheral T-cell lymphoma, not otherwise 3 4 Seong Jin Cho · In Sun Kim specified (PTCL-NOS), and 13 cases of extranodal NK/T-cell lymphoma, nasal type (NKTCL). Thus, we attempted to elucidate the associations of the degree of the expression of histone acetyltrans- Department of Pathology, Hallym University ferase 1 (HAT1), histone deacetylase (HDAC) 1, HDAC2, and HDAC3 with the clinical behaviors of Sacred Heart Hospital, Hallym University College above malignant lymphomas using the immunohistochemistry and a western blot analysis. of Medicine; 1Ilsong Institute of Life Science, Re- Hallym University; 2Department of Hemato- sults: The degree of the expression of HAT1 was higher in cases of DLBCL, PTCL-NOS or NKT- Oncology, Hallym University Sacred Heart CL as compared with reactive lymphoid hyperplasia (p< 0.05). The degree of the expression of Hospital, Hallym University College of Medicine, HAT1 was correlated with that of HDAC1 in cases of DLBCL or NKTCL (p< 0.05).
    [Show full text]
  • The Tumor Suppressor ING5 Is a Dimeric, Bivalent Recognition Molecule of the Histone H3k4me3 Mark
    Article The Tumor Suppressor ING5 Is a Dimeric, Bivalent Recognition Molecule of the Histone H3K4me3 Mark Georgina Ormaza 1,†, Jhon A. Rodríguez 1,†, Alain Ibáñez de Opakua 1, Nekane Merino 1, Maider Villate 1, Irantzu Gorroño 1, Miriam Rábano 1, Ignacio Palmero 2, Marta Vilaseca 3, Robert Kypta 1,4, María d.M. Vivanco 1, Adriana L. Rojas 1 and Francisco J. Blanco 1,5 1 - CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain 2 - Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC-UAM, 28029 Madrid, Spain 3 - Institute for Research in Biomedicine, 08028 Barcelona, Spain 4 - Department of Surgery and Cancer, Imperial College London, London, W12 0NN, UK 5 - IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain Correspondence to Francisco J. Blanco: CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain. [email protected] https://doi.org/10.1016/j.jmb.2019.04.018 Edited by M. Guss Abstract The INhibitor of Growth (ING) family of tumor suppressors regulates the transcriptional state of chromatin by recruiting remodeling complexes to sites with histone H3 trimethylated at lysine 4 (H3K4me3). This modification is recognized by the plant homeodomain (PHD) present at the C-terminus of the five ING proteins. ING5 facilitates histone H3 acetylation by the HBO1 complex, and also H4 acetylation by the MOZ/ MORF complex. We show that ING5 forms homodimers through its N-terminal domain, which folds independently into an elongated coiled-coil structure. The central region of ING5, which contains the nuclear localization sequence, is flexible and disordered, but it binds dsDNA with micromolar affinity. NMR analysis of the full-length protein reveals that the two PHD fingers of the dimer are chemically equivalent and independent of the rest of the molecule, and they bind H3K4me3 in the same way as the isolated PHD.
    [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]
  • Cancer TNT Ashwin Ram 12/5/2017 Background: Chromatin Writers, Readers, Erasers
    Cancer TNT Ashwin Ram 12/5/2017 Background: Chromatin Writers, Readers, Erasers writer effector eg. HAT, HMT reader eg. bromodomain eraser eg. HDAC, KDM The writer HAT1: A known H4 lysine 5,12 di-acetyltransferase writer siHAT1 siControl HAT1 H4 K12Ac H4 K5Ac actin Western blot for histone H4 modifications after control and HAT1 siRNA transfections. HAT1: EGF-stimulated immunoprecipitation specific specific - - HAT1 IgG HAT1 IgG - - R α non R α non EGF: + + + - - - WB: HAT1 Immunoprecipitation / WB to measure HAT1 levels +/- Heatmap of gene expression changes of all human histone EGF acetyltransferases +/- EGF and siRNA treatments shows HAT1 expression is EGF-dependent Working model of HAT1 : The oldest “new” histone acetyltransferase EGF EGFR plasma membrane HAT1 H4 H3 Rbap46/48 a nuclear membrane H4 H2A H3 H2B HAT1 S phase Surprise: HAT1 also binds (a few sites) on chromatin HAT1 ChipSeq signal sits on Hist1 locus on Chromosome 6 Read Depth HAT1 bound sites (zoom) HAT1 ChIP-seq peaks cluster at Read Depth histone H4 promoters. Hist1H2BE Hist1H4D Hist1H3D Hist1H4E Is HAT1 a transcription factor for its substrate (H4)? EGF EGFR plasma membrane HAT1 H4 H3 Rbap46/48 a nuclear membrane H4 H2A H3 H2B HAT1 S phase HAT1 is required for S-phase burst of histone H4 mRNA HAT1 Hist1H4B mRNA level 50 Rbap46 45 shCont-3 H4 40 shHAT1-A7 35 shHAT1-B6 30 25 B6 A7 - - 20 15 shHAT1 shHAT1 shControl 10 Gene actin)Expression (versus 5 HAT1 0 0 2 4 6 8 10 actin hours after release from double thymidine block G1 S G2/M HAT1 loss: Life with less histones EGF
    [Show full text]
  • Recombinant Human HAT1 Protein Catalog Number: ATGP0507
    Recombinant human HAT1 protein Catalog Number: ATGP0507 PRODUCT INPORMATION Expression system E.coli Domain 20-341aa UniProt No. O14929 NCBI Accession No. NP_003633 Alternative Names Histone acetyltransferase 1, KAT1, Histone acetyltransferase 1 HAT 1, Histidine aminotransferase 1, Histone acetyltransferase type B catalytic subunit PRODUCT SPECIFICATION Molecular Weight 40.1 kDa (343aa) confirmed by MALDI-TOF Concentration 0.5mg/ml (determined by Bradford assay) Formulation Liquid in. 20mM Tris-HCl buffer (pH 8.0) containing 10% glycerol, 1mM DTT Purity > 90% by SDS-PAGE Tag His-Tag Application SDS-PAGE Storage Condition Can be stored at +2C to +8C for 1 week. For long term storage, aliquot and store at -20C to -80C. Avoid repeated freezing and thawing cycles. BACKGROUND Description HAT1, also known as histone acetyltransferase 1, is a type B histone acetyltransferase (HAT) that is involved in the rapid acetylation of newly synthesized cytoplasmic histones, which are in turn imported into the nucleus for de novo deposition onto nascent DNA chains. Histone acetylation, particularly of histone H4, plays an important role in replication-dependent chromatin assembly. Specifically, this HAT can acetylate soluble but not nucleosomal histone H4 at lysines 5 and 12, and to a lesser degree, histone H2A at lysine 5. Recombinant HAT1 1 Recombinant human HAT1 protein Catalog Number: ATGP0507 protein was expressed in E. coli and purified by using conventional chromatography techniques. Amino acid Sequence MGSSHHHHHH SSGLVPRGSH MKKLAEYKCN TNTAIELKLV RFPEDLENDI RTFFPEYTHQ LFGDDETAFG YKGLKILLYY IAGSLSTMFR VEYASKVDEN FDCVEADDVE GKIRQIIPPG FCTNTNDFLS LLEKEVDFKP FGTLLHTYSV LSPTGGENFT FQIYKADMTC RGFREYHERL QTFLMWFIET ASFIDVDDER WHYFLVFEKY NKDGATLFAT VGYMTVYNYY VYPDKTRPRV SQMLILTPFQ GQGHGAQLLE TVHRYYTEFP TVLDITAEDP SKSYVKLRDF VLVKLCQDLP CFSREKLMQG FNEDMAIEAQ QKFKINKQHA RRVYEILRLL VTD General References Verreault A., et al.
    [Show full text]
  • HAT1 Sirna (Rat)
    For research purposes only, not for human use Product Data Sheet HAT1 siRNA (Rat) Catalog # Source Reactivity Applications CRR4463 Synthetic R RNAi Description siRNA to inhibit HAT1 expression using RNA interference Specificity HAT1 siRNA (Rat) is a target-specific 19-23 nt siRNA oligo duplexes designed to knock down gene expression. Form Lyophilized powder Gene Symbol HAT1 Alternative Names Histone acetyltransferase type B catalytic subunit; Histone acetyltransferase 1 Entrez Gene 296501 (Rat) SwissProt Q5M939 (Rat) Purity > 97% Quality Control Oligonucleotide synthesis is monitored base by base through trityl analysis to ensure appropriate coupling efficiency. The oligo is subsequently purified by affinity-solid phase extraction. The annealed RNA duplex is further analyzed by mass spectrometry to verify the exact composition of the duplex. Each lot is compared to the previous lot by mass spectrometry to ensure maximum lot-to-lot consistency. Components We offers pre-designed sets of 3 different target-specific siRNA oligo duplexes of rat HAT1 gene. Each vial contains 5 nmol of lyophilized siRNA. The duplexes can be transfected individually or pooled together to achieve knockdown of the target gene, which is most commonly assessed by qPCR or western blot. Our siRNA oligos are also chemically modified (2’-OMe) at no extra charge for increased stability and enhanced knockdown in vitro and in vivo. Directions for Use We recommends transfection with 100 nM siRNA 48 to 72 hours prior to cell lysis. Application key: E- ELISA, WB- Western blot,
    [Show full text]
  • The Role of ING5 in Maintaining Stemness of Brain Tumor Initiating Cells
    University of Calgary PRISM: University of Calgary's Digital Repository Graduate Studies The Vault: Electronic Theses and Dissertations 2016 The Role of ING5 in Maintaining Stemness of Brain Tumor Initiating Cells Wang, Fangwu Jr Wang, F. J. (2016). The Role of ING5 in Maintaining Stemness of Brain Tumor Initiating Cells (Unpublished master's thesis). University of Calgary, Calgary, AB. doi:10.11575/PRISM/28327 http://hdl.handle.net/11023/3233 master thesis University of Calgary graduate students retain copyright ownership and moral rights for their thesis. You may use this material in any way that is permitted by the Copyright Act or through licensing that has been assigned to the document. For uses that are not allowable under copyright legislation or licensing, you are required to seek permission. Downloaded from PRISM: https://prism.ucalgary.ca UNIVERSITY OF CALGARY The Role of ING5 in Maintaining Stemness of Brain Tumor Initiating Cells by Fangwu Wang A THESIS SUBMITTED TO THE FACULTY OF GRADUATE STUDIES IN PARTIAL FULFILMENT OF THE REQUIREMENTS FOR THE DEGREE OF MASTER OF SCIENCE GRADUATE PROGRAM IN BIOCHEMISTRY AND MOLECULAR BIOLOGY CALGARY, ALBERTA AUGUST, 2016 © Fangwu Wang 2016 Abstract Brain tumor initiating cells (BTICs) are believed to account for the recurrence of glioblastomas following treatment. Recent studies have shown that the stemness of BTICs and intratumoral differentiation hierarchy are determined largely on the epigenetic level. The ING family of epigenetic regulators function in diverse growth regulatory, metastasis and chemoresistance pathways, through targeting different histone acetyltransferase (HAT) and histone deacetylase (HDAC) complexes to the H3K4me3 mark to alter histone acetylation.
    [Show full text]
  • Table S6. Names and Functions of 44 Target Genes and 4 Housekeeping Genes Assessed for Gene Expression Measurements
    Table S6. Names and functions of 44 target genes and 4 housekeeping genes assessed for gene expression measurements. Gene Gene name Function Category Reference CD45 CD45 (leukocyte common antigen) Regulation of T-cell and B-cell antigen receptor signaling Adaptive NCBI, UniProt HIVEP2 Human immunodeficiency virus typeI enhancer2 Transcription factor, V(D)J recombination, MHC enhancer binding Adaptive (Diepeveen et al. 2013) HIVEP3 Human immunodeficiency virus typeI enhancer3 Transcription factor, V(D)J recombination, MHC enhancer binding Adaptive (Diepeveen et al. 2013) IgM-lc Immunoglobulin light chain Recognition of antigen or pathogen Adaptive NCBI, UniProt Integ-Bt Integrin-beta 1 Cell signaling and adhesion of immunoglobulin Adaptive NCBI, UniProt Lymph75 Lymphocyte antigen 75 Directs captured antigens to lymphocytes Adaptive (Birrer et al. 2012) Lympcyt Lymphocyte cytosolic protein 2 Positive role in promoting T-cell development and activation Adaptive NCBI, UniProt TAP Tap-binding protein (Tapasin) Transport of antigenic peptides, peptide loading on MHC I Adaptive NCBI, UniProt AIF Allograft inflammation factor Inflammatory responses, allograft rejection, activation of macrophages Innate (Roth et al. 2012) Calrcul Calreticulin Chaperone, promotes phagocytosis and clearance of apoptotic cells Innate NCBI, UniProt Cf Coagulation factor II Blood clotting and inflammation response Innate (Birrer et al. 2012) IL8 Interleukin 8 Neutrophil chemotactic factor, phagocytosis, inflammatory activity Innate NCBI, UniProt Intf Interferon induced transmembrane protein 3 Negative regulation of viral entry into host cell, antiviral response Innate NCBI, UniProt Kin Kinesin Intracellular transport Innate (Roth et al. 2012) LectptI Lectin protein type I Pathogen recognition receptors (C-type lectin type I) Innate NCBI, UniProt LectpII Lectin protein type II Pathogen recognition receptors (C-type lectin type II) Innate NCBI, UniProt Nramp Natural resistance-assoc macrophage protein Macrophage activation Innate (Roth et al.
    [Show full text]
  • Hat2p Recognizes the Histone H3 Tail to Specify the Acetylation of the Newly Synthesized H3/H4 Heterodimer by the Hat1p/Hat2p Complex
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press Hat2p recognizes the histone H3 tail to specify the acetylation of the newly synthesized H3/H4 heterodimer by the Hat1p/Hat2p complex Yang Li,1,4 Li Zhang,1,4 Tingting Liu,1,4 Chengliang Chai,1,4 Qianglin Fang,2 Han Wu,1 Paula A. Agudelo Garcia,3 Zhifu Han,1 Shuai Zong,1 You Yu,1 Xinyue Zhang,1 Mark R. Parthun,3 Jijie Chai,1 Rui-Ming Xu,2 and Maojun Yang1,5 1Ministry of Education Key Laboratory of Protein Sciences, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; 2National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; 3Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, Ohio 43210, USA Post-translational modifications of histones are significant regulators of replication, transcription, and DNA repair. Particularly, newly synthesized histone H4 in H3/H4 heterodimers becomes acetylated on N-terminal lysine residues prior to its incorporation into chromatin. Previous studies have established that the histone acetyltransferase (HAT) complex Hat1p/Hat2p medicates this modification. However, the mechanism of how Hat1p/Hat2p recognizes and facilitates the enzymatic activities on the newly assembled H3/H4 heterodimer remains unknown. Furthermore, Hat2p is a WD40 repeat protein, which is found in many histone modifier complexes. However, how the WD40 repeat proteins facilitate enzymatic activities of histone modification enzymes is unclear. In this study, we first solved the high-resolution crystal structure of a Hat1p/Hat2p/CoA/H4 peptide complex and found that the H4 tail interacts with both Hat1p and Hat2p, by which substrate recruitment is facilitated.
    [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]
  • Open Data for Differential Network Analysis in Glioma
    International Journal of Molecular Sciences Article Open Data for Differential Network Analysis in Glioma , Claire Jean-Quartier * y , Fleur Jeanquartier y and Andreas Holzinger Holzinger Group HCI-KDD, Institute for Medical Informatics, Statistics and Documentation, Medical University Graz, Auenbruggerplatz 2/V, 8036 Graz, Austria; [email protected] (F.J.); [email protected] (A.H.) * Correspondence: [email protected] These authors contributed equally to this work. y Received: 27 October 2019; Accepted: 3 January 2020; Published: 15 January 2020 Abstract: The complexity of cancer diseases demands bioinformatic techniques and translational research based on big data and personalized medicine. Open data enables researchers to accelerate cancer studies, save resources and foster collaboration. Several tools and programming approaches are available for analyzing data, including annotation, clustering, comparison and extrapolation, merging, enrichment, functional association and statistics. We exploit openly available data via cancer gene expression analysis, we apply refinement as well as enrichment analysis via gene ontology and conclude with graph-based visualization of involved protein interaction networks as a basis for signaling. The different databases allowed for the construction of huge networks or specified ones consisting of high-confidence interactions only. Several genes associated to glioma were isolated via a network analysis from top hub nodes as well as from an outlier analysis. The latter approach highlights a mitogen-activated protein kinase next to a member of histondeacetylases and a protein phosphatase as genes uncommonly associated with glioma. Cluster analysis from top hub nodes lists several identified glioma-associated gene products to function within protein complexes, including epidermal growth factors as well as cell cycle proteins or RAS proto-oncogenes.
    [Show full text]