The Putative Tumor Suppressors EXT1 and EXT2 Form a Stable Complex That Accumulates in the Golgi Apparatus and Catalyzes the Synthesis of Heparan Sulfate

Total Page:16

File Type:pdf, Size:1020Kb

The Putative Tumor Suppressors EXT1 and EXT2 Form a Stable Complex That Accumulates in the Golgi Apparatus and Catalyzes the Synthesis of Heparan Sulfate The putative tumor suppressors EXT1 and EXT2 form a stable complex that accumulates in the Golgi apparatus and catalyzes the synthesis of heparan sulfate Craig McCormick, Gillian Duncan, K. Tina Goutsos, and Frank Tufaro* Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada Edited by Kai Simons, European Molecular Biology Laboratory, Heidelberg, Germany, and approved November 19, 1999 (received for review August 23, 1999) Hereditary multiple exostoses, a dominantly inherited genetic raised by these observations. EXT1, when overexpressed in a disorder characterized by multiple cartilaginous tumors, is caused cell, appears to be localized predominantly to the ER (17, 21), by mutations in members of the EXT gene family, EXT1 or EXT2. The whereas the biosynthesis of HS chains is thought to occur in the proteins encoded by these genes, EXT1 and EXT2, are endoplasmic Golgi cisternae (22–25). Moreover, if the EXT1 and EXT2 genes reticulum-localized type II transmembrane glycoproteins that pos- encode functionally redundant HS polymerases (HS-Pol), it is sess or are tightly associated with glycosyltransferase activities not clear why mutations in either gene cause HME. involved in the polymerization of heparan sulfate. Here, by testing To address these questions, we overexpressed functional a cell line with a specific defect in EXT1 in in vivo and in vitro assays, epitope-tagged and native forms of EXT1 and EXT2 in cells and we show that EXT2 does not harbor significant glycosyltransferase examined their subcellular localization and enzymatic activity. activity in the absence of EXT1. Instead, it appears that EXT1 and By using a cell line, sog9, with a specific defect in EXT1, we show EXT2 form a hetero-oligomeric complex in vivo that leads to the that EXT2 does not harbor significant glycosyltransferase activ- accumulation of both proteins in the Golgi apparatus. Remarkably, ity in the absence of EXT1. Instead, it appears that EXT1 and the Golgi-localized EXT1͞EXT2 complex possesses substantially EXT2 form a hetero-oligomeric complex in vivo that leads to an higher glycosyltransferase activity than EXT1 or EXT2 alone, which accumulation of both proteins in the Golgi apparatus. Remark- suggests that the complex represents the biologically relevant ably, the Golgi-localized EXT1͞EXT2 complex possesses sub- form of the enzyme(s). These findings provide a rationale to stantially higher glycosyltransferase activity than EXT1 or EXT2 explain how inherited mutations in either of the two EXT genes can alone, which suggests that this complex represents the biologi- cause loss of activity, resulting in hereditary multiple exostoses. cally relevant form of the enzyme(s). These findings provide a rationale to explain how inherited mutations in either of the two ereditary multiple exostoses (HME) is an autosomal dom- EXT genes can cause loss of activity, resulting in hereditary Hinant disorder characterized by the formation of cartilage- multiple exostoses. capped tumors (exostoses) that develop from the growth plate of Materials and Methods endochondral bone (1). This condition can lead to skeletal EXT Constructs. pEXT1 was isolated from a HeLa cell cDNA abnormalities, short stature, and in some instances, malignant library in pcDNA3.1 (A550–26, Invitrogen) as described previ- transformation from exostoses to chondrosarcomas (2, 3) or ously (17). pEXT1 myc-His, pG339DEXT1 myc-His, and osteosarcomas (4, 5). Although genetic linkage analysis has pR340CEXT1 myc-His were constructed as described previously identified three different loci for HME, EXT1 on 8q24.1, EXT2 (17). All reagents were obtained from Life Technologies unless on 11p11–13, and EXT3 on 19p (6–8), most HME cases have otherwise stated. pEXT1 GFP was constructed by excision of been attributed to missense or frameshift mutations in either EXT1 from pEXT1 myc-His with BamHI and SstII, followed by EXT1 or EXT2 (9–15). EXT1 and EXT2 encode 746- and 718-aa ligation into the BglII and SstII sites in the pEGFP-N1 expression proteins, respectively, that are expressed ubiquitously in human vector (CLONTECH). The bovine EXT2 constructs were con- tissues (9, 16). structed as previously described (18). pbEXT2 myc was con- Previous studies using epitope-tagged constructs have dem- structed by PCR of the EXT2 coding region by using primers onstrated that EXT1 is a predominantly endoplasmic reticulum 5Ј-CGG GAT CCC GGT TTC ATT ATG TGT GCG TCA GTC (ER)-localized glycoprotein whose expression enhances the AAG TCC AAC A-3Ј and 5Ј-GCT CTA GAG CTC ACA GAT synthesis of cell surface heparan sulfate (HS) (17). HS chains are CCT CTT CTG AGA TGA GTT TTT GTT CTA AGC TGC composed of alternating residues of D-glucuronic acid (GlcA) Ј 3 CAA TGT TGG-3 . After digestion with BamHI and XbaI, the and N-acetyl-D-glucosamine (GlcNAc) joined by 1 4 linkages, bEXT2 myc PCR product was then ligated into pcDNA3.1͞myc- and a recent study has shown that both EXT1 and EXT2 harbor His B. A murine EXT2 (mEXT2) cDNA was a gift from M. GlcA transferase (GlcA-T) and GlcNAc transferase (GlcNAc-T) Lovett (Washington University School of Medicine, St. Louis). activities that catalyze the polymerization of HS (18). EXT1 and EXT2 are structurally similar to previously identified glycosyl- transferases in that they are type II transmembrane proteins This paper was submitted directly (Track II) to the PNAS office. comprising an N-terminal cytoplasmic tail, a transmembrane Abbreviations: HME, hereditary multiple exostoses; ER, endoplasmic reticulum; HS, hepa- domain, a stalk, and a large globular domain that is likely to ran sulfate; GlcA, D-glucuronic acid; GlcNAc, N-acetyl-D-glucosamine; GlcA-T, glucuronyl- harbor enzymatic activity (19). Moreover, a truncated active transferase; GlcNAc-T, N-acetyl-D-glucosaminyltransferase; HS-Pol, heparan sulfate poly- merase; HSV-1, herpes simplex virus type I; GFP, green fluorescent protein; endoH, en- form of EXT2 is secreted from cells and can be isolated from doglycosidase H. serum (18), which is a fate common to other ER and Golgi- *To whom reprint requests should be addressed. E-mail: [email protected]. localized glycosyltransferases, including EXTL2, which is an ␣ The publication costs of this article were defrayed in part by page charge payment. This EXT homolog shown to encode an 1,4-N-acetylhexosaminyl- article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. transferase (20). However, several important questions are §1734 solely to indicate this fact. 668–673 ͉ PNAS ͉ January 18, 2000 ͉ vol. 97 ͉ no. 2 Downloaded by guest on September 26, 2021 pmEXT2 GFP was constructed by PCR using primers 5Ј-CGG Immunoprecipitations. BHK cells (1 ϫ 106) were transfected with GAT CCC GGT TTC ATT ATG TGT GCG TCA GTC AAG green fluorescent protein (GFP) or Myc-His-tagged EXT con- TCC AAC A-3Ј and 5Ј-TCC CCG CGG GGA TAA GCT GCC structs. After 20 h, cells were radiolabeled with 100 ␮Ci͞ml (1 AAT GTT GGG GAA-3Ј. The mEXT2 PCR product was ␮Ci ϭ 37 kBq) [35S]methionine (ICN) in methionine- and ligated into T-tailed pBluescript (Stratagene), followed by di- cysteine-free DMEM (ICN) for 1.5 h at 37°C. Cells were washed gestion with HindIII and EcoRI and subsequent ligation into with PBS and lysed in Triton lysis buffer [2% Triton X-100͞20 ⅐ ͞ pEGFP-N1. pmEXT2 myc-His was constructed by digestion of mM Tris HCl pH 7.4 150 mM NaCl containing CØmplete pmEXT2 GFP with HindIII and SstII and ligation into the protease inhibitors (Roche)] at 4°C for 15 min. The lysates were ϫ pcDNA3.1͞myc-His B vector. To isolate human EXTL2, centrifuged at 12,000 g for 15 min, and precleared for 30 min ␮ EXTL3, and murine N-deacetylase͞N-sulfotransferase-2 with 25 l of protein G-Sepharose (Pharmacia) at 4°C. The lysates were then incubated with 0.5 ␮g of mouse anti-Myc (NDST2), total cellular RNA was isolated from confluent HeLa ␮ cell and L cell cultures by using Trizol reagent, reverse tran- monoclonal antibody (Invitrogen) or 0.5 g of rabbit anti-GFP scribed and amplified by PCR using primers 5Ј-CCG CTC GAG monoclonal antibody (CLONTECH) for 2 h, followed by incu- Ј Ј bation with 25 ␮l of protein G-Sepharose for 1 h. The lysates CGG AAT TAA ACT TCA ACA CAA TG-3 and 5 -GGG ϫ Ј were centrifuged at 12,000 g for 10 s, and washed two times GTA CCC CTA TTT TTC TTT TGT AGT TGG CAT-3 for ⅐ ͞ ͞ ͞ Ј with 10 mM Tris HCl pH 7.4 150 mM NaCl 2 mM EDTA 0.2% EXTL2,5-CCG CTC GAG CGG CAG GCT GCA GAG GAC ⅐ ͞ Ј Ј Triton X-100, two times with 10 mM Tris HCl, pH 7.4 500 mM TCA T-3 and 5 -GGG GTA CCC CGA TGA ACT TGA AGC ͞ ͞ Ј Ј NaCl 2 mM EDTA 0.2% Triton X-100, and two times with 10 ACT TGG TCT-3 for EXTL3 and 5 -GAA GAT CTT CCC mM Tris⅐HCl, pH 7.4. The pellet was suspended in 30 ␮lof ACC ATG CTC CAG CTG TGG AAG GT-3Ј and 5Ј-CGG ͞ ͞ Ј SDS PAGE sample buffer and boiled for 5 min before SDS AAT TCC GCC CAC ACT GGA ATG TTG CAA T-3 for PAGE. Proteins were transferred to Immobilon-P membranes NDST2. The digested PCR products were ligated into the (Millipore) and exposed to BioMAX MR film (Kodak). appropriate sites in pEGFP-N1. G339DEXT1-GFP, ⌬ R340CEXT1-GFP, and NTMEXT1-GFP were constructed by Assay of Cellular Glycosyltransferase Activities. BHK or mutant digestion and subsequent ligation into pEGFP-N1. sog9 cells were transfected with EXT constructs. At 30 h after transfection, cells were washed in PBS and lysed in Triton͞ Herpes Simplex Virus Type 1 (HSV-1) Infection Assay.
Recommended publications
  • CRISPR Screening of Porcine Sgrna Library Identifies Host Factors
    ARTICLE https://doi.org/10.1038/s41467-020-18936-1 OPEN CRISPR screening of porcine sgRNA library identifies host factors associated with Japanese encephalitis virus replication Changzhi Zhao1,5, Hailong Liu1,5, Tianhe Xiao1,5, Zichang Wang1, Xiongwei Nie1, Xinyun Li1,2, Ping Qian2,3, Liuxing Qin3, Xiaosong Han1, Jinfu Zhang1, Jinxue Ruan1, Mengjin Zhu1,2, Yi-Liang Miao 1,2, Bo Zuo1,2, ✉ ✉ Kui Yang4, Shengsong Xie 1,2 & Shuhong Zhao 1,2 1234567890():,; Japanese encephalitis virus (JEV) is a mosquito-borne zoonotic flavivirus that causes ence- phalitis and reproductive disorders in mammalian species. However, the host factors critical for its entry, replication, and assembly are poorly understood. Here, we design a porcine genome-scale CRISPR/Cas9 knockout (PigGeCKO) library containing 85,674 single guide RNAs targeting 17,743 protein-coding genes, 11,053 long ncRNAs, and 551 microRNAs. Subsequently, we use the PigGeCKO library to identify key host factors facilitating JEV infection in porcine cells. Several previously unreported genes required for JEV infection are highly enriched post-JEV selection. We conduct follow-up studies to verify the dependency of JEV on these genes, and identify functional contributions for six of the many candidate JEV- related host genes, including EMC3 and CALR. Additionally, we identify that four genes associated with heparan sulfate proteoglycans (HSPGs) metabolism, specifically those responsible for HSPGs sulfurylation, facilitate JEV entry into porcine cells. Thus, beyond our development of the largest CRISPR-based functional genomic screening platform for pig research to date, this study identifies multiple potentially vulnerable targets for the devel- opment of medical and breeding technologies to treat and prevent diseases caused by JEV.
    [Show full text]
  • Promiscuity and Specificity of Eukaryotic Glycosyltransferases
    Biochemical Society Transactions (2020) 48 891–900 https://doi.org/10.1042/BST20190651 Review Article Promiscuity and specificity of eukaryotic glycosyltransferases Ansuman Biswas and Mukund Thattai Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, TIFR, Bangalore, India Correspondence: Mukund Thattai ([email protected]) Glycosyltransferases are a large family of enzymes responsible for covalently linking sugar monosaccharides to a variety of organic substrates. These enzymes drive the synthesis of complex oligosaccharides known as glycans, which play key roles in inter-cellular interac- tions across all the kingdoms of life; they also catalyze sugar attachment during the syn- thesis of small-molecule metabolites such as plant flavonoids. A given glycosyltransferase enzyme is typically responsible for attaching a specific donor monosaccharide, via a spe- cific glycosidic linkage, to a specific moiety on the acceptor substrate. However these enzymes are often promiscuous, able catalyze linkages between a variety of donors and acceptors. In this review we discuss distinct classes of glycosyltransferase promiscuity, each illustrated by enzymatic examples from small-molecule or glycan synthesis. We high- light the physical causes of promiscuity, and its biochemical consequences. Structural studies of glycosyltransferases involved in glycan synthesis show that they make specific contacts with ‘recognition motifs’ that are much smaller than the full oligosaccharide sub- strate. There is a wide range in the sizes of glycosyltransferase recognition motifs: highly promiscuous enzymes recognize monosaccharide or disaccharide motifs across multiple oligosaccharides, while highly specific enzymes recognize large, complex motifs found on few oligosaccharides. In eukaryotes, the localization of glycosyltransferases within compartments of the Golgi apparatus may play a role in mitigating the glycan variability caused by enzyme promiscuity.
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • NICU Gene List Generator.Xlsx
    Neonatal Crisis Sequencing Panel Gene List Genes: A2ML1 - B3GLCT A2ML1 ADAMTS9 ALG1 ARHGEF15 AAAS ADAMTSL2 ALG11 ARHGEF9 AARS1 ADAR ALG12 ARID1A AARS2 ADARB1 ALG13 ARID1B ABAT ADCY6 ALG14 ARID2 ABCA12 ADD3 ALG2 ARL13B ABCA3 ADGRG1 ALG3 ARL6 ABCA4 ADGRV1 ALG6 ARMC9 ABCB11 ADK ALG8 ARPC1B ABCB4 ADNP ALG9 ARSA ABCC6 ADPRS ALK ARSL ABCC8 ADSL ALMS1 ARX ABCC9 AEBP1 ALOX12B ASAH1 ABCD1 AFF3 ALOXE3 ASCC1 ABCD3 AFF4 ALPK3 ASH1L ABCD4 AFG3L2 ALPL ASL ABHD5 AGA ALS2 ASNS ACAD8 AGK ALX3 ASPA ACAD9 AGL ALX4 ASPM ACADM AGPS AMELX ASS1 ACADS AGRN AMER1 ASXL1 ACADSB AGT AMH ASXL3 ACADVL AGTPBP1 AMHR2 ATAD1 ACAN AGTR1 AMN ATL1 ACAT1 AGXT AMPD2 ATM ACE AHCY AMT ATP1A1 ACO2 AHDC1 ANK1 ATP1A2 ACOX1 AHI1 ANK2 ATP1A3 ACP5 AIFM1 ANKH ATP2A1 ACSF3 AIMP1 ANKLE2 ATP5F1A ACTA1 AIMP2 ANKRD11 ATP5F1D ACTA2 AIRE ANKRD26 ATP5F1E ACTB AKAP9 ANTXR2 ATP6V0A2 ACTC1 AKR1D1 AP1S2 ATP6V1B1 ACTG1 AKT2 AP2S1 ATP7A ACTG2 AKT3 AP3B1 ATP8A2 ACTL6B ALAS2 AP3B2 ATP8B1 ACTN1 ALB AP4B1 ATPAF2 ACTN2 ALDH18A1 AP4M1 ATR ACTN4 ALDH1A3 AP4S1 ATRX ACVR1 ALDH3A2 APC AUH ACVRL1 ALDH4A1 APTX AVPR2 ACY1 ALDH5A1 AR B3GALNT2 ADA ALDH6A1 ARFGEF2 B3GALT6 ADAMTS13 ALDH7A1 ARG1 B3GAT3 ADAMTS2 ALDOB ARHGAP31 B3GLCT Updated: 03/15/2021; v.3.6 1 Neonatal Crisis Sequencing Panel Gene List Genes: B4GALT1 - COL11A2 B4GALT1 C1QBP CD3G CHKB B4GALT7 C3 CD40LG CHMP1A B4GAT1 CA2 CD59 CHRNA1 B9D1 CA5A CD70 CHRNB1 B9D2 CACNA1A CD96 CHRND BAAT CACNA1C CDAN1 CHRNE BBIP1 CACNA1D CDC42 CHRNG BBS1 CACNA1E CDH1 CHST14 BBS10 CACNA1F CDH2 CHST3 BBS12 CACNA1G CDK10 CHUK BBS2 CACNA2D2 CDK13 CILK1 BBS4 CACNB2 CDK5RAP2
    [Show full text]
  • SUPPLEMENTARY MATERIAL Effect of Next
    SUPPLEMENTARY MATERIAL Effect of Next-Generation Exome Sequencing Depth for Discovery of Diagnostic Variants KKyung Kim1,2,3†, Moon-Woo Seong4†, Won-Hyong Chung3, Sung Sup Park4, Sangseob Leem1, Won Park5,6, Jihyun Kim1,2, KiYoung Lee1,2*‡, Rae Woong Park1,2* and Namshin Kim5,6** 1Department of Biomedical Informatics, Ajou University School of Medicine, Suwon 443-749, Korea 2Department of Biomedical Science, Graduate School, Ajou University, Suwon 443-749, Korea, 3Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Korea, 4Department of Laboratory Medicine, Seoul National University Hospital College of Medicine, Seoul 110-799, Korea, 5Department of Functional Genomics, Korea University of Science and Technology, Daejeon 305-806, Korea, 6Epigenomics Research Center, Genome Institute, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Korea http//www. genominfo.org/src/sm/gni-13-31-s001.pdf Supplementary Table 1. List of diagnostic genes Gene Symbol Description Associated diseases ABCB11 ATP-binding cassette, sub-family B (MDR/TAP), member 11 Intrahepatic cholestasis ABCD1 ATP-binding cassette, sub-family D (ALD), member 1 Adrenoleukodystrophy ACVR1 Activin A receptor, type I Fibrodysplasia ossificans progressiva AGL Amylo-alpha-1, 6-glucosidase, 4-alpha-glucanotransferase Glycogen storage disease ALB Albumin Analbuminaemia APC Adenomatous polyposis coli Adenomatous polyposis coli APOE Apolipoprotein E Apolipoprotein E deficiency AR Androgen receptor Androgen insensitivity
    [Show full text]
  • Viruses Like Sugars: How to Assess Glycan Involvement in Viral Attachment
    microorganisms Review Viruses Like Sugars: How to Assess Glycan Involvement in Viral Attachment Gregory Mathez and Valeria Cagno * Institute of Microbiology, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; [email protected] * Correspondence: [email protected] Abstract: The first step of viral infection requires interaction with the host cell. Before finding the specific receptor that triggers entry, the majority of viruses interact with the glycocalyx. Identifying the carbohydrates that are specifically recognized by different viruses is important both for assessing the cellular tropism and for identifying new antiviral targets. Advances in the tools available for studying glycan–protein interactions have made it possible to identify them more rapidly; however, it is important to recognize the limitations of these methods in order to draw relevant conclusions. Here, we review different techniques: genetic screening, glycan arrays, enzymatic and pharmacological approaches, and surface plasmon resonance. We then detail the glycan interactions of enterovirus D68 and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), highlighting the aspects that need further clarification. Keywords: attachment receptor; viruses; glycan; sialic acid; heparan sulfate; HBGA; SARS-CoV-2; EV-D68 Citation: Mathez, G.; Cagno, V. Viruses Like Sugars: How to Assess 1. Introduction Glycan Involvement in Viral This review focuses on methods for assessing the involvement of carbohydrates in Attachment. Microorganisms 2021, 9, viral attachment and entry into the host cell. Viruses often bind to entry receptors that are 1238. https://doi.org/10.3390/ not abundant on the cell surface; to increase their chances of finding them, they initially microorganisms9061238 bind to attachment receptors comprising carbohydrates that are more widely expressed.
    [Show full text]
  • Supplementary Materials and Tables a and B
    SUPPLEMENTARY MATERIAL 1 Table A. Main characteristics of the subset of 23 AML patients studied by high-density arrays (subset A) WBC BM blasts MYST3- MLL Age/Gender WHO / FAB subtype Karyotype FLT3-ITD NPM status (x109/L) (%) CREBBP status 1 51 / F M4 NA 21 78 + - G A 2 28 / M M4 t(8;16)(p11;p13) 8 92 + - G G 3 53 / F M4 t(8;16)(p11;p13) 27 96 + NA G NA 4 24 / M PML-RARα / M3 t(15;17) 5 90 - - G G 5 52 / M PML-RARα / M3 t(15;17) 1.5 75 - - G G 6 31 / F PML-RARα / M3 t(15;17) 3.2 89 - - G G 7 23 / M RUNX1-RUNX1T1 / M2 t(8;21) 38 34 - + ND G 8 52 / M RUNX1-RUNX1T1 / M2 t(8;21) 8 68 - - ND G 9 40 / M RUNX1-RUNX1T1 / M2 t(8;21) 5.1 54 - - ND G 10 63 / M CBFβ-MYH11 / M4 inv(16) 297 80 - - ND G 11 63 / M CBFβ-MYH11 / M4 inv(16) 7 74 - - ND G 12 59 / M CBFβ-MYH11 / M0 t(16;16) 108 94 - - ND G 13 41 / F MLLT3-MLL / M5 t(9;11) 51 90 - + G R 14 38 / F M5 46, XX 36 79 - + G G 15 76 / M M4 46 XY, der(10) 21 90 - - G NA 16 59 / M M4 NA 29 59 - - M G 17 26 / M M5 46, XY 295 92 - + G G 18 62 / F M5 NA 67 88 - + M A 19 47 / F M5 del(11q23) 17 78 - + M G 20 50 / F M5 46, XX 61 59 - + M G 21 28 / F M5 46, XX 132 90 - + G G 22 30 / F AML-MD / M5 46, XX 6 79 - + M G 23 64 / M AML-MD / M1 46, XY 17 83 - + M G WBC: white blood cell.
    [Show full text]
  • Epigenetic Mechanisms Are Involved in the Oncogenic Properties of ZNF518B in Colorectal Cancer
    Epigenetic mechanisms are involved in the oncogenic properties of ZNF518B in colorectal cancer Francisco Gimeno-Valiente, Ángela L. Riffo-Campos, Luis Torres, Noelia Tarazona, Valentina Gambardella, Andrés Cervantes, Gerardo López-Rodas, Luis Franco and Josefa Castillo SUPPLEMENTARY METHODS 1. Selection of genomic sequences for ChIP analysis To select the sequences for ChIP analysis in the five putative target genes, namely, PADI3, ZDHHC2, RGS4, EFNA5 and KAT2B, the genomic region corresponding to the gene was downloaded from Ensembl. Then, zoom was applied to see in detail the promoter, enhancers and regulatory sequences. The details for HCT116 cells were then recovered and the target sequences for factor binding examined. Obviously, there are not data for ZNF518B, but special attention was paid to the target sequences of other zinc-finger containing factors. Finally, the regions that may putatively bind ZNF518B were selected and primers defining amplicons spanning such sequences were searched out. Supplementary Figure S3 gives the location of the amplicons used in each gene. 2. Obtaining the raw data and generating the BAM files for in silico analysis of the effects of EHMT2 and EZH2 silencing The data of siEZH2 (SRR6384524), siG9a (SRR6384526) and siNon-target (SRR6384521) in HCT116 cell line, were downloaded from SRA (Bioproject PRJNA422822, https://www.ncbi. nlm.nih.gov/bioproject/), using SRA-tolkit (https://ncbi.github.io/sra-tools/). All data correspond to RNAseq single end. doBasics = TRUE doAll = FALSE $ fastq-dump -I --split-files SRR6384524 Data quality was checked using the software fastqc (https://www.bioinformatics.babraham. ac.uk /projects/fastqc/). The first low quality removing nucleotides were removed using FASTX- Toolkit (http://hannonlab.cshl.edu/fastxtoolkit/).
    [Show full text]
  • Glycan Metabolism a Validated Grna Library for CRISPR/Cas9
    Glycobiology, 2018, vol. 28, no. 5, 295–305 doi: 10.1093/glycob/cwx101 Advance Access Publication Date: 5 January 2018 Original Article Glycan Metabolism A validated gRNA library for CRISPR/Cas9 targeting of the human glycosyltransferase Downloaded from https://academic.oup.com/glycob/article-abstract/28/5/295/4791732 by guest on 08 October 2018 genome Yoshiki Narimatsu2,3,1, Hiren J Joshi2, Zhang Yang2,3, Catarina Gomes2,4, Yen-Hsi Chen2, Flaminia C Lorenzetti 2, Sanae Furukawa2, Katrine T Schjoldager2, Lars Hansen2, Henrik Clausen2, Eric P Bennett2,1, and Hans H Wandall2 2Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark, 3GlycoDisplay Aps, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark, and 4Instituto de Investigação e Inovação em Saúde,i3S; Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal 1To whom correspondence should be addressed: Tel: +45-35335528; Fax: +45-35367980; e-mail: [email protected] (Y.N.); Tel: +4535326630; Fax: +45-35367980; e-mail: [email protected] (E.P.B.) Received 25 September 2017; Revised 20 November 2017; Editorial decision 5 December 2017; Accepted 7 December 2017 Abstract Over 200 glycosyltransferases are involved in the orchestration of the biosynthesis of the human glycome, which is comprised of all glycan structures found on different glycoconjugates in cells. The glycome is vast, and despite advancements in analytic strategies it continues to be difficult to decipher biological roles of glycans with respect to specific glycan structures, type of glycoconju- gate, particular glycoproteins, and distinct glycosites on proteins.
    [Show full text]
  • Hereditary Multiple Exostoses (EXT1 & EXT2 Concurrently) Sequencing
    Lab Dept: Anatomic Pathology Test Name: HEREDITARY MULTIPLE EXOSTOSES (EXT1 & EXT2 CONCURRENTLY) SEQUENCING General Information Lab Order Codes: EX12 Synonyms: Exostoses 1 and 2; Hereditary multiple osteochondromata; Multiple cartilaginous exostoses; Diaphyseal aclasis; HME CPT Codes: 81479 x4 – Molecular Pathology Unlisted procedure Test Includes: Using genomic DNA obtained from the submitted specimen, bi-directional sequence of all 11 coding exons of the EXT1 gene is obtained and analyzed. Concurrently, targeted array CGH analysis with exon-level resolution (ExonArrayDx) is performed to evaluate for a deletion or duplication of one or more exons in both the EXT1 and EXT2 genes. Mutations found in the first person of a family to be tested are confirmed by repeat analysis using sequencing, restriction fragment analysis, or other appropriate method. Logistics Test Indications: Individuals with HME often develop benign cartilage-capped tumors (exostoses) at the ends of the long bones or the surface of flat bones. Exostoses develop prior to skeletal maturity only. Bony deformity, bowing of the long bones, limited range of motion, and premature osteoarthrosis may be associated with hereditary multiple exostoses (HME). Exostoses also may cause complications by putting pressure on nearby tissues, nerves or blood vessels. A rare but severe risk in patients with mutiples exostoses is the development of malignant chondrosarcoma, which in 1-5% of patients. Mutation in the EXT1 gene seems to be associated with a more severe disease and higher risk of developing chondrosarcoma than EXT2 mutations. Reasons for referral: 1. Confirmation of a clinical diagnosis 2. Genetic counseling Lab Testing Sections: Anatomic Pathology - Sendouts Referred to: GeneDx, Inc.
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2003/0198970 A1 Roberts (43) Pub
    US 2003O19897OA1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2003/0198970 A1 Roberts (43) Pub. Date: Oct. 23, 2003 (54) GENOSTICS clinical trials on groups or cohorts of patients. This group data is used to derive a Standardised method of treatment (75) Inventor: Gareth Wyn Roberts, Cambs (GB) which is Subsequently applied on an individual basis. There is considerable evidence that a significant factor underlying Correspondence Address: the individual variability in response to disease, therapy and FINNEGAN, HENDERSON, FARABOW, prognosis lies in a person's genetic make-up. There have GARRETT & DUNNER been numerous examples relating that polymorphisms LLP within a given gene can alter the functionality of the protein 1300 ISTREET, NW encoded by that gene thus leading to a variable physiological WASHINGTON, DC 20005 (US) response. In order to bring about the integration of genomics into medical practice and enable design and building of a (73) Assignee: GENOSTIC PHARMA LIMITED technology platform which will enable the everyday practice (21) Appl. No.: 10/206,568 of molecular medicine a way must be invented for the DNA Sequence data to be aligned with the identification of genes (22) Filed: Jul. 29, 2002 central to the induction, development, progression and out come of disease or physiological States of interest. Accord Related U.S. Application Data ing to the invention, the number of genes and their configu rations (mutations and polymorphisms) needed to be (63) Continuation of application No. 09/325,123, filed on identified in order to provide critical clinical information Jun. 3, 1999, now abandoned. concerning individual prognosis is considerably less than the 100,000 thought to comprise the human genome.
    [Show full text]
  • SSIEM Classification of Inborn Errors of Metabolism 2011
    SSIEM classification of Inborn Errors of Metabolism 2011 Disease group / disease ICD10 OMIM 1. Disorders of amino acid and peptide metabolism 1.1. Urea cycle disorders and inherited hyperammonaemias 1.1.1. Carbamoylphosphate synthetase I deficiency 237300 1.1.2. N-Acetylglutamate synthetase deficiency 237310 1.1.3. Ornithine transcarbamylase deficiency 311250 S Ornithine carbamoyltransferase deficiency 1.1.4. Citrullinaemia type1 215700 S Argininosuccinate synthetase deficiency 1.1.5. Argininosuccinic aciduria 207900 S Argininosuccinate lyase deficiency 1.1.6. Argininaemia 207800 S Arginase I deficiency 1.1.7. HHH syndrome 238970 S Hyperammonaemia-hyperornithinaemia-homocitrullinuria syndrome S Mitochondrial ornithine transporter (ORNT1) deficiency 1.1.8. Citrullinemia Type 2 603859 S Aspartate glutamate carrier deficiency ( SLC25A13) S Citrin deficiency 1.1.9. Hyperinsulinemic hypoglycemia and hyperammonemia caused by 138130 activating mutations in the GLUD1 gene 1.1.10. Other disorders of the urea cycle 238970 1.1.11. Unspecified hyperammonaemia 238970 1.2. Organic acidurias 1.2.1. Glutaric aciduria 1.2.1.1. Glutaric aciduria type I 231670 S Glutaryl-CoA dehydrogenase deficiency 1.2.1.2. Glutaric aciduria type III 231690 1.2.2. Propionic aciduria E711 232000 S Propionyl-CoA-Carboxylase deficiency 1.2.3. Methylmalonic aciduria E711 251000 1.2.3.1. Methylmalonyl-CoA mutase deficiency 1.2.3.2. Methylmalonyl-CoA epimerase deficiency 251120 1.2.3.3. Methylmalonic aciduria, unspecified 1.2.4. Isovaleric aciduria E711 243500 S Isovaleryl-CoA dehydrogenase deficiency 1.2.5. Methylcrotonylglycinuria E744 210200 S Methylcrotonyl-CoA carboxylase deficiency 1.2.6. Methylglutaconic aciduria E712 250950 1.2.6.1. Methylglutaconic aciduria type I E712 250950 S 3-Methylglutaconyl-CoA hydratase deficiency 1.2.6.2.
    [Show full text]