Inactivating Mutations and X-Ray Crystal Structure of the Tumor Suppressor OPCML Reveal Cancer-Associated Functions

Total Page:16

File Type:pdf, Size:1020Kb

Inactivating Mutations and X-Ray Crystal Structure of the Tumor Suppressor OPCML Reveal Cancer-Associated Functions ARTICLE https://doi.org/10.1038/s41467-019-10966-8 OPEN Inactivating mutations and X-ray crystal structure of the tumor suppressor OPCML reveal cancer-associated functions James R. Birtley1,6,7, Mohammad Alomary2,7, Elisa Zanini2, Jane Antony 2, Zachary Maben1, Grant C. Weaver 1, Claudia Von Arx2, Manuela Mura2, Aline T. Marinho2, Haonan Lu 2, Eloise V.N. Morecroft2,3, Evdoxia Karali2, Naomi E. Chayen 4, Edward W. Tate 3, Mollie Jurewicz1, Lawrence J. Stern 1,8, Chiara Recchi 2,8 & Hani Gabra 2,5,8 1234567890():,; OPCML, a tumor suppressor gene, is frequently silenced epigenetically in ovarian and other cancers. Here we report, by analysis of databases of tumor sequences, the observation of OPCML somatic missense mutations from various tumor types and the impact of these mutations on OPCML function, by solving the X-ray crystal structure of this glycoprotein to 2.65 Å resolution. OPCML consists of an extended arrangement of three immunoglobulin-like domains and homodimerizes via a network of contacts between membrane-distal domains. We report the generation of a panel of OPCML variants with representative clinical mutations and demonstrate clear phenotypic effects in vitro and in vivo including changes to anchorage- independent growth, interaction with activated cognate receptor tyrosine kinases, cellular migration, invasion in vitro and tumor growth in vivo. Our results suggest that clinically occurring somatic missense mutations in OPCML have the potential to contribute to tumorigenesis in a variety of cancers. 1 Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA. 2 Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, Du Cane Road, London W12 0NN, UK. 3 Department of Chemistry, Imperial College London, Wood Lane, London W12 0BZ, UK. 4 Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK. 5 Clinical Discovery Unit, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Cambridge SG8 6HB, UK. 6Present address: UCB Pharma, Bath Road, Slough SL1 3WE, UK. 7These authors contributed equally: James R. Birtley, Mohammad Alomary. 8These authors jointly supervised this work: Lawrence J. Stern, Chiara Recchi, Hani Gabra. Correspondence and requests for materials should be addressed to L.J.S. (email: lawrence. [email protected]) or to C.R. (email: [email protected]) or to H.G. (email: [email protected]) NATURE COMMUNICATIONS | (2019) 10:3134 | https://doi.org/10.1038/s41467-019-10966-8 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-10966-8 pioid-binding protein cell-adhesion molecule like allowed for chain tracing, which was followed by rounds of (OPCML) is a glycosylphosphatidylinositol (GPI)- automated and manual model building and refinement (see O fl anchored protein that localizes to the outer lea et of the Methods). In the final model (PDB entry 5UV6), 273 out of 286 plasma membrane and acts as a tumor suppressor (TS)1,2. residues in the mature OPCML protein were visualized (Sup- OPCML is a member of the IgLON superfamily, together with 4 plementary Figs. 2 and 3), with two molecules in the crystal- other proteins (LSAMP, NEGR1, HNT, and IgLON5), amongst lographic asymmetric unit. There are six potential N-linked which some have been hypothesized to act as tumor suppressors glycosylation sites (asparagines 44, 70, 140, 285, 293, and 306) or oncogenes3,4. However, functions of IgLON members have not and glycans were modelled at residues 70, 293, and 306. been fully characterized and their 3D structures remain unsolved. OPCML consists of three Ig-like domains termed D1, D2, and Previously, we showed that OPCML is inactivated predominantly D3, connected by short extended linkers (Fig. 1a, b, Supplemen- by somatic methylation and loss of heterozygosity in more than tary Fig. 2). The OPCML homodimer resembles an inverted V 80% of ovarian cancer patients2. When re-expressed in cancer shape (Fig. 1a), with dimerization mediated by a membrane-distal cells, OPCML inhibits proliferation in vitro and tumorigenicity D1-D1 interface stabilized by a combination of hydrophobic in vivo2 by binding and downregulating a specific subset of contacts, hydrogen bonds and salt bridges (Fig. 1c, Supplemen- receptor tyrosine kinases (RTKs)5. Although somatic promoter tary Fig. 4). This interface comprises 874 Å2 of buried surface area methylation is the predominant mechanism of silencing in and has a solvation free energy gain (ΔiG) on formation diverse tumor types1, we previously also reported a somatic calculated by PISA to be −9.5 kcal M−1. These values are larger missense mutation at P95R in an ovarian cancer patient2. Pre- than for other intermolecular interaction sites observed in the liminary data indicated that this variant was expressed in similar crystal, the next largest being crystal contacts mediated by the C- amounts as wild-type (WT) but was a loss-of-function mutant. terminal end of one molecule inserting between the B and G Here, we search for additional OPCML somatic mutations in the strands of D1 in another with 554 Å2 buried surface area and TCGA and COSMIC DNA sequencing databases from a panel of solvation free energy gain of −6.9 kcal M−1 (Supplementary nearly 30,000 patients affected by different cancers. We show that table 2 and Supplementary Fig. 5). Based on the quaternary a number of patients exhibit somatic mutations of OPCML.In structure seen in the asymmetric unit, the dimer would be order to understand how these clinical mutations might interfere anchored into the plasma membrane by two D3-linked GPI with OPCML’s tumor suppressor activity we then determine the anchors (Fig. 1a). The D3 C-terminal ends are located crystal structure of OPCML and show that these mutations fall approximately 180 Åapart, and thus the top of the D1 into several classes based on mutation type and location on the dimerization interface could extend above the plane of the protein structure. Selecting mutations representative of each class, plasma membrane by ~73 Å (Fig. 1a). we characterize their biophysical properties and demonstrate The OPCML dimer is stabilized by reciprocal salt bridges their functionality in in vitro and in vivo models. We further between Arg 65 of one monomer and Asp 80 of the other (Fig. 1c, demonstrate that particular mutations affect the oligomeric state of OPCML, its interaction with cognate RTKs and its effect on a range of phenotypes including migration, invasion, adhesion to extracellular matrix proteins and in vivo tumorigenicity. These fi fi Table 1 Data collection, phasing, and re nement statistics ndings reveal how the mutations result in functional/clinical for OPCML (MR-SAD) consequences, and these data have implications for under- standing the mechanism of action of this class of membrane- Native K2PtCl4 associated tumor suppressors. Data collection Space group P41212I4122 Cell dimensions Results a, b, c (Å) 93.6, 93.6, 262.2 98.8, 98.8, 267.1 OPCML sustains point mutations in cancer patients.Weana- α, β, γ (°) 90, 90, 90 90, 90, 90 a lyzed tumor DNA sequence data from the TCGA and COSMIC Resolution (Å) 18.5–2.65 (2.74–2.65) 48.3–3.17 (3.25–3.17) databases in order to identify possible clinical OPCML mutations. Rsym 17.1 (273.6) 18.7 (301.5) We found point mutations in the OPCML gene in 287 out of 28,132 Rpim 4.7 (73.8) 3.6 (56.6) patients and these were distributed across all cancer types (Sup- I/σ(I) 9.7 (1.2) 11.5 (1.1) CC1/2 100 (68.5) 100 (82.3) plementary Fig. 1A). Generally, the frequency of these clinical Completeness (%) 95.8 (88.8) 100.0 (100.0) mutations is uncommon, ranging from 5.5% in melanoma and Redundancy 14.4 (14.5) 28.2 (29.1) 3.3% in colon, gastric, and bladder cancer to around 0.1% in cancers Refinement Resolution (Å) 18.5–2.65 (2.74–2.65) such as breast and brain (Supplementary Fig. 1A). Nine mutations No. reflections 33374 (3021) were found to be splice site variants or stop codon-related altera- Rwork/Rfree 28.7/30.4 tions (Supplementary Table 1). The remaining 278 changes were No. atoms 4392 Protein 4216 missense mutations (Supplementary Table 1) and were mostly Ligand/ion (carbohydrate) 134 scattered evenly along the entire OPCML sequence, with the Water 42 greatest number located in domain 1 (Supplementary Fig. 1B). B factors Protein 84.6 Ligand/ion 91.0 Water 74.2 X-ray crystal structure of OPCML. To localize the clinical R.m.s deviations mutations on the three-dimensional structure of OPCML and to Bond lengths (Å) 0.006 gain insights into their role in the tumor suppressor mechanism(s) Bond angles (°) 0.99 we crystallized soluble recombinant OPCML (residues 36–316) NCS RMSD (Å) All domains 1.10 and determined its structure using a combined single-wavelength D1 (43–134) 0.24 X-ray anomalous dispersion/molecular replacement approach D2 (135–222) 0.19 (Table 1). Structure solution was complicated initially by a high D3 (223–319) 0.64 fi degree of translational pseudosymmetry, but identi cation of the aValues in parentheses are for highest-resolution shell correct space group and non-crystallographic symmetry elements 2 NATURE COMMUNICATIONS | (2019) 10:3134 | https://doi.org/10.1038/s41467-019-10966-8 | www.nature.com/naturecommunications NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-019-10966-8 ARTICLE a * * c e D1 D1 Arg 65 Thr 114 WT Leu 69 Dimer structure Asp 80 Trp 82 N N Ile 74 1 Monomer structure D2 D2 ~73Å Ile 74 D3 D3 Trp 82 Leu 69 Thr 114 Asp 80 Arg 65 Scaled intensity (log RU) 0.00 0.05 0.10 0.15 0.20 q (Å–1) C WT fit to dimer Membrane 1.5 WT fit to monomer d 1.0 Thr 94 ~180Å Val 92 Asn 93 Pro 95 0.5 0 3.1 Å 3.0 Å 0.00 0.05 0.10 0.15 0.20 b 3.2 Å Ratio of intensity (RU) q (Å–1) 2.9 Å C * Gln 97 Thr 96 WT OPCML Ser 99 f Tyr 98 C ) (arbitrary units) R ( * p 0 50 100 150 200 R (Å) Fig.
Recommended publications
  • Hearing Aging Is 14.1±0.4% GWAS-Heritable
    medRxiv preprint doi: https://doi.org/10.1101/2021.07.05.21260048; this version posted July 7, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-NC 4.0 International license . Predicting age from hearing test results with machine learning reveals the genetic and environmental factors underlying accelerated auditory aging Alan Le Goallec1,2+, Samuel Diai1+, Théo Vincent1, Chirag J. Patel1* 1Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA 2Department of Systems, Synthetic and Quantitative Biology, Harvard University, Cambridge, MA, 02118, USA +Co-first authors *Corresponding author Contact information: Chirag J Patel [email protected] Abstract With the aging of the world population, age-related hearing loss (presbycusis) and other hearing disorders such as tinnitus become more prevalent, leading to reduced quality of life and social isolation. Unveiling the genetic and environmental factors leading to age-related auditory disorders could suggest lifestyle and therapeutic interventions to slow auditory aging. In the following, we built the first machine learning-based hearing age predictor by training models to predict chronological age from hearing test results (root mean squared error=7.10±0.07 years; R-Squared=31.4±0.8%). We defined hearing age as the prediction outputted by the model on unseen samples, and accelerated auditory aging as the difference between a participant’s hearing age and age. We then performed a genome wide association study [GWAS] and found that accelerated hearing aging is 14.1±0.4% GWAS-heritable.
    [Show full text]
  • Serum Cell-Free DNA Methylation of OPCML and HOXD9 As a Biomarker That May Aid in Differential Diagnosis Between Cholangiocarcin
    Wasenang et al. Clinical Epigenetics (2019) 11:39 https://doi.org/10.1186/s13148-019-0634-0 RESEARCH Open Access Serum cell-free DNA methylation of OPCML and HOXD9 as a biomarker that may aid in differential diagnosis between cholangiocarcinoma and other biliary diseases Wiphawan Wasenang1,2, Ponlatham Chaiyarit3, Siriporn Proungvitaya1,4 and Temduang Limpaiboon1,4* Abstract Background: Cholangiocarcinoma (CCA) is a fatal cancer of the bile duct epithelial cell lining. The misdiagnosis of CCA and other biliary diseases may occur due to the similarity of clinical manifestations and blood tests resulting in inappropriate or delayed treatment. Thus, an accurate and less-invasive method for differentiating CCA from other biliary diseases is inevitable. Methods: We quantified methylation of OPCML, HOXA9,andHOXD9 in serum cell-free DNA (cfDNA) of CCA patients and other biliary diseases using methylation-sensitive high-resolution melting (MS-HRM). Their potency as differential biomarkers between CCA and other biliary diseases was also evaluated by using receiver operating characteristic (ROC) curves. Results: The significant difference of methylation levels of OPCML and HOXD9 was observed in serum cfDNA of CCA compared to other biliary diseases. Assessment of serum cfDNA methylation of OPCML and HOXD9 as differential biomarkers of CCA and other biliary diseases showed the area under curve (AUC) of 0.850 (0.759–0.941) for OPCML which sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and accuracy were 80.00%, 90.00%, 88.88%, 81.81%, and 85.00%, respectively. The AUC of HOXD9 was 0.789 (0.686–0.892) with sensitivity, specificity, PPV, NPV, and accuracy of 67.50%, 90.00%, 87.09%, 73.46%, and 78.75%, respectively.
    [Show full text]
  • Supplementary Table 1: Adhesion Genes Data Set
    Supplementary Table 1: Adhesion genes data set PROBE Entrez Gene ID Celera Gene ID Gene_Symbol Gene_Name 160832 1 hCG201364.3 A1BG alpha-1-B glycoprotein 223658 1 hCG201364.3 A1BG alpha-1-B glycoprotein 212988 102 hCG40040.3 ADAM10 ADAM metallopeptidase domain 10 133411 4185 hCG28232.2 ADAM11 ADAM metallopeptidase domain 11 110695 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 195222 8038 hCG40937.4 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 165344 8751 hCG20021.3 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 189065 6868 null ADAM17 ADAM metallopeptidase domain 17 (tumor necrosis factor, alpha, converting enzyme) 108119 8728 hCG15398.4 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 117763 8748 hCG20675.3 ADAM20 ADAM metallopeptidase domain 20 126448 8747 hCG1785634.2 ADAM21 ADAM metallopeptidase domain 21 208981 8747 hCG1785634.2|hCG2042897 ADAM21 ADAM metallopeptidase domain 21 180903 53616 hCG17212.4 ADAM22 ADAM metallopeptidase domain 22 177272 8745 hCG1811623.1 ADAM23 ADAM metallopeptidase domain 23 102384 10863 hCG1818505.1 ADAM28 ADAM metallopeptidase domain 28 119968 11086 hCG1786734.2 ADAM29 ADAM metallopeptidase domain 29 205542 11085 hCG1997196.1 ADAM30 ADAM metallopeptidase domain 30 148417 80332 hCG39255.4 ADAM33 ADAM metallopeptidase domain 33 140492 8756 hCG1789002.2 ADAM7 ADAM metallopeptidase domain 7 122603 101 hCG1816947.1 ADAM8 ADAM metallopeptidase domain 8 183965 8754 hCG1996391 ADAM9 ADAM metallopeptidase domain 9 (meltrin gamma) 129974 27299 hCG15447.3 ADAMDEC1 ADAM-like,
    [Show full text]
  • Recombinant Human FLRT2 Protein Catalog Number: ATGP3291
    Recombinant human FLRT2 protein Catalog Number: ATGP3291 PRODUCT INPORMATION Expression system Baculovirus Domain 36-541aa UniProt No. O43155 NCBI Accession No. NP_037363 Alternative Names FLRT2 PRODUCT SPECIFICATION Molecular Weight 57.5 kDa (514aa) Concentration 0.25mg/ml (determined by absorbance at 280nm) Formulation Liquid in. Phosphate-Buffered Saline (pH 7.4) containing 10% glycerol Purity > 90% by SDS-PAGE Endotoxin level < 1 EU per 1ug of protein (determined by LAL method) 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 FLRT2, also known as leucine-rich repeat transmembrane protein FLRT2, is one of three FLRT (fibronectin, leucine rich repeat, transmembrane) glycoproteins expressed in distinct areas of the developing brain and other tissues. Human FLRT1 and FLRT3 ECDs (extracellular domain) share approximately 47% aa identity with FLRT2. The fibronectin domain of all three FLRTs can bind to FGF receptors. Recombinant human FLRT2, fused to His-tag 1 Recombinant human FLRT2 protein Catalog Number: ATGP3291 at C-terminus, was expressed in insect cell and purified by using conventional chromatography techniques. Amino acid Sequence CPSVCRCDRN FVYCNERSLT SVPLGIPEGV TVLYLHNNQI NNAGFPAELH NVQSVHTVYL YGNQLDEFPM NLPKNVRVLH LQENNIQTIS RAALAQLLKL EELHLDDNSI STVGVEDGAF REAISLKLLF LSKNHLSSVP VGLPVDLQEL RVDENRIAVI SDMAFQNLTS LERLIVDGNL LTNKGIAEGT FSHLTKLKEF SIVRNSLSHP PPDLPGTHLI RLYLQDNQIN HIPLTAFSNL RKLERLDISN NQLRMLTQGV FDNLSNLKQL TARNNPWFCD CSIKWVTEWL KYIPSSLNVR GFMCQGPEQV RGMAVRELNM NLLSCPTTTP GLPLFTPAPS TASPTTQPPT LSIPNPSRSY TPPTPTTSKL PTIPDWDGRE RVTPPISERI QLSIHFVNDT SIQVSWLSLF TVMAYKLTWV KMGHSLVGGI VQERIVSGEK QHLSLVNLEP RSTYRICLVP LDAFNYRAVE DTICSEATTH ASYLNNGSNT ASSHEQTTSH SMGSPFLEHH HHHH General References Haines B.P., et al. (2006) Dev.
    [Show full text]
  • Recombinant Human Fibronectin Leucine Rich Transmembrane Protein 2/FLRT2 (C-6His)
    9853 Pacific Heights Blvd. Suite D. San Diego, CA 92121, USA Tel: 858-263-4982 Email: [email protected] 32-7320: Recombinant Human Fibronectin Leucine Rich Transmembrane Protein 2/FLRT2 (C-6His) Gene : FLRT2 Gene ID : 23768 Uniprot ID : O43155 Description Source: Human Cells. MW :57.3kD. Recombinant Human FLRT2 is produced by our Mammalian expression system and the target gene encoding Cys36-Ser539 is expressed with a 6His tag at the C-terminus. Fibronectin Leucine Rich Transmembrane protein 2 (FLRT2) is a member of the fibronectin leucine rich transmembrane protein (FLRT) family. The three fibronectin leucine-rich repeat transmembrane (FLRT) proteins: FLRT1, FLRT2 and FLRT3, all contain 10 leucine-rich repeats (LRR), a type III fibronectin (FN) domain, followed by the transmembrane region, and a short cytoplasmic tail. FLRT proteins have dual properties as regulators of cell adhesion and potentiators of fibroblast growth factor (FGF) mediated signalling. The fibronectin domain of all three FLRTs can bind FGF receptors. This binding is thought to regulate FGF signaling during development. The LRR domains are responsible for both the localization of FLRTs in areas of cell contact and homotypic cell cell association. FLRT2 is expressed in a subset of the sclerotome, adjacent to the region that forms the syndetome, suggesting its involvement in the FGF signalling pathway. Product Info Amount : 10 µg / 50 µg Content : Lyophilized from a 0.2 µm filtered solution of 20mM PB, 150mM NaCl, pH 7.2. Lyophilized protein should be stored at -20°C, though stable at room temperature for 3 weeks. Storage condition : Reconstituted protein solution can be stored at 4-7°C for 2-7 days.
    [Show full text]
  • Role and Regulation of the P53-Homolog P73 in the Transformation of Normal Human Fibroblasts
    Role and regulation of the p53-homolog p73 in the transformation of normal human fibroblasts Dissertation zur Erlangung des naturwissenschaftlichen Doktorgrades der Bayerischen Julius-Maximilians-Universität Würzburg vorgelegt von Lars Hofmann aus Aschaffenburg Würzburg 2007 Eingereicht am Mitglieder der Promotionskommission: Vorsitzender: Prof. Dr. Dr. Martin J. Müller Gutachter: Prof. Dr. Michael P. Schön Gutachter : Prof. Dr. Georg Krohne Tag des Promotionskolloquiums: Doktorurkunde ausgehändigt am Erklärung Hiermit erkläre ich, dass ich die vorliegende Arbeit selbständig angefertigt und keine anderen als die angegebenen Hilfsmittel und Quellen verwendet habe. Diese Arbeit wurde weder in gleicher noch in ähnlicher Form in einem anderen Prüfungsverfahren vorgelegt. Ich habe früher, außer den mit dem Zulassungsgesuch urkundlichen Graden, keine weiteren akademischen Grade erworben und zu erwerben gesucht. Würzburg, Lars Hofmann Content SUMMARY ................................................................................................................ IV ZUSAMMENFASSUNG ............................................................................................. V 1. INTRODUCTION ................................................................................................. 1 1.1. Molecular basics of cancer .......................................................................................... 1 1.2. Early research on tumorigenesis ................................................................................. 3 1.3. Developing
    [Show full text]
  • Shear Stress Modulates Gene Expression in Normal Human Dermal Fibroblasts
    University of Calgary PRISM: University of Calgary's Digital Repository Graduate Studies The Vault: Electronic Theses and Dissertations 2017 Shear Stress Modulates Gene Expression in Normal Human Dermal Fibroblasts Zabinyakov, Nikita Zabinyakov, N. (2017). Shear Stress Modulates Gene Expression in Normal Human Dermal Fibroblasts (Unpublished master's thesis). University of Calgary, Calgary, AB. doi:10.11575/PRISM/27775 http://hdl.handle.net/11023/3639 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 Shear Stress Modulates Gene Expression in Normal Human Dermal Fibroblasts by Nikita Zabinyakov 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 BIOMEDICAL ENGINEERING CALGARY, ALBERTA JANUARY 2017 © Nikita Zabinyakov 2017 Abstract Applied mechanical forces, such as those resulting from fluid flow, trigger cells to change their functional behavior or phenotype. However, there is little known about how fluid flow affects fibroblasts. The hypothesis of this thesis is that dermal fibroblasts undergo significant changes of expression of differentiation genes after exposure to fluid flow (or shear stress). To test the hypothesis, human dermal fibroblasts were exposed to laminar steady fluid flow for 20 and 40 hours and RNA was collected for microarray analysis.
    [Show full text]
  • FLRT Proteins Act As Guidance Cues for Migrating Cortical Interneurons
    Nom/Logotip de la Universitat on s’ha llegit la tesi FLRT proteins act as guidance cues for migrating cortical interneurons Catherine Fleitas Pérez http://hdl.handle.net/10803/378646 ADVERTIMENT. L'accés als continguts d'aquesta tesi doctoral i la seva utilització ha de respectar els drets de la persona autora. Pot ser utilitzada per a consulta o estudi personal, així com en activitats o materials d'investigació i docència en els termes establerts a l'art. 32 del Text Refós de la Llei de Propietat Intel·lectual (RDL 1/1996). Per altres utilitzacions es requereix l'autorització prèvia i expressa de la persona autora. En qualsevol cas, en la utilització dels seus continguts caldrà indicar de forma clara el nom i cognoms de la persona autora i el títol de la tesi doctoral. No s'autoritza la seva reproducció o altres formes d'explotació efectuades amb finalitats de lucre ni la seva comunicació pública des d'un lloc aliè al servei TDX. Tampoc s'autoritza la presentació del seu contingut en una finestra o marc aliè a TDX (framing). Aquesta reserva de drets afecta tant als continguts de la tesi com als seus resums i índexs. ADVERTENCIA. El acceso a los contenidos de esta tesis doctoral y su utilización debe respetar los derechos de la persona autora. Puede ser utilizada para consulta o estudio personal, así como en actividades o materiales de investigación y docencia en los términos establecidos en el art. 32 del Texto Refundido de la Ley de Propiedad Intelectual (RDL 1/1996). Para otros usos se requiere la autorización previa y expresa de la persona autora.
    [Show full text]
  • Bioinformatics Tools for the Analysis of Gene-Phenotype Relationships Coupled with a Next Generation Chip-Sequencing Data Processing Pipeline
    Bioinformatics Tools for the Analysis of Gene-Phenotype Relationships Coupled with a Next Generation ChIP-Sequencing Data Processing Pipeline Erinija Pranckeviciene Thesis submitted to the Faculty of Graduate and Postdoctoral Studies in partial fulfillment of the requirements for the Doctorate in Philosophy degree in Cellular and Molecular Medicine Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa c Erinija Pranckeviciene, Ottawa, Canada, 2015 Abstract The rapidly advancing high-throughput and next generation sequencing technologies facilitate deeper insights into the molecular mechanisms underlying the expression of phenotypes in living organisms. Experimental data and scientific publications following this technological advance- ment have rapidly accumulated in public databases. Meaningful analysis of currently avail- able data in genomic databases requires sophisticated computational tools and algorithms, and presents considerable challenges to molecular biologists without specialized training in bioinfor- matics. To study their phenotype of interest molecular biologists must prioritize large lists of poorly characterized genes generated in high-throughput experiments. To date, prioritization tools have primarily been designed to work with phenotypes of human diseases as defined by the genes known to be associated with those diseases. There is therefore a need for more prioritiza- tion tools for phenotypes which are not related with diseases generally or diseases with which no genes have yet been associated in particular. Chromatin immunoprecipitation followed by next generation sequencing (ChIP-Seq) is a method of choice to study the gene regulation processes responsible for the expression of cellular phenotypes. Among publicly available computational pipelines for the processing of ChIP-Seq data, there is a lack of tools for the downstream analysis of composite motifs and preferred binding distances of the DNA binding proteins.
    [Show full text]
  • Transcriptomic Causal Networks Identified Patterns of Differential Gene Regulation in Human Brain from Schizophrenia Cases Versus Controls
    Transcriptomic Causal Networks identified patterns of differential gene regulation in human brain from Schizophrenia cases versus controls Akram Yazdani1, Raul Mendez-Giraldez2, Michael R Kosorok3, Panos Roussos1,4,5 1Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA 2Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, NC, USA 3Department of Biostatistics, University of North Carolina at Chapel Hill, NC, USA 4Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA 5Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, New York, 10468, USA Abstract Common and complex traits are the consequence of the interaction and regulation of multiple genes simultaneously, which work in a coordinated way. However, the vast majority of studies focus on the differential expression of one individual gene at a time. Here, we aim to provide insight into the underlying relationships of the genes expressed in the human brain in cases with schizophrenia (SCZ) and controls. We introduced a novel approach to identify differential gene regulatory patterns and identify a set of essential genes in the brain tissue. Our method integrates genetic, transcriptomic, and Hi-C data and generates a transcriptomic-causal network. Employing this approach for analysis of RNA-seq data from CommonMind Consortium, we identified differential regulatory patterns for SCZ cases and control groups to unveil the mechanisms that control the transcription of the genes in the human brain. Our analysis identified modules with a high number of SCZ-associated genes as well as assessing the relationship of the hubs with their down-stream genes in both, cases and controls.
    [Show full text]
  • Genetic Analysis of FLRT Protein Family During Early Mouse Embryonic Development
    Genetic analysis of the FLRT family of proteins during early mouse embryonic development Dissertation Der Fakultät für Biologie der Ludwig-Maximilians-Universität München Eingereicht am 17. Februar 2009 von Christian Erlacher 1. Gutachter: Prof. Dr. Rüdiger Klein 2. Gutachter: PD Dr. Angelika Böttger Tag der mündlichen Prüfung: 24.04.2009 Work presented in this dissertation was performed in the laboratory of Prof. Dr. Rüdiger Klein, Department of Molecular Neurobiology, Max Planck Institute of Neurobiology, Martinsried, Germany. Work was performed under the supervision of Prof. Dr. Rüdiger Klein and Dr. Joaquim E g e a Ehrenwörtliche Versicherung Ich versichere hiermit ehrenwörtlich, dass die vorgelegte Dissertation von mir selbständig und ohne unerlaubte Beihilfe angefertigt ist. München, den (Unterschrift) Erklärung Hiermit erkläre ich, dass ich mich anderweitig einer Doktorprüfung ohne Erfolg nicht unterzogen habe. München, den (Unterschrift) Publication from the work presented in this dissertation # Joaquim E g e a* , Christian Erlacher*, Eloi Montanez, Ingo Burtscher, Satoru Yamagishi, Martin Heß, Falko Hampel, Rodrigo Sanchez, Maria Teresa Rodriguez-Manzaneque, Michael R. Bösl, Reinhard Fässler, Heiko Lickert, and Rüdiger Klein# „Genetic ablation of FLRT3 reveals a novel morphogenetic function for the anterior visceral endoderm in suppressing mesoderm differentiation.“ Genes & Development 22, Nr. 23 (Dec 2008): 3349-3362. * and #: These authors contributed equally to this work. Für Margareta und Hans Table of contents Abbreviations V List of Figures IX 1. Introduction 1 1.1. Preimplantation development 3 1.2. Postimplantation development 4 1.3. Axis specification 8 1.4. Molecular basis of axis formation 11 1.5. Gastrulation 15 1.6. Molecular mechanisms guiding gastrulation 18 1.7.
    [Show full text]
  • Down-Regulated Expression of OPCML Predicts an Unfavorable
    Xing et al. BMC Cancer (2017) 17:268 DOI 10.1186/s12885-017-3203-y RESEARCHARTICLE Open Access Down-regulated expression of OPCML predicts an unfavorable prognosis and promotes disease progression in human gastric cancer Xiangbin Xing1*†, Weibin Cai2†, Sanmei Ma3†, Yongfei Wang3, Huijuan Shi4, Ming Li1,3, Jinxia Jiao1,3, Yang Yang1,3, Longshan Liu5, Xiangliang Zhang6 and Minhu Chen1* Abstract Background: OPCML belongs to the IgLON family of Ig domain–containing GPI-anchored cell adhesion molecules and was recently found to be involved in carcinogenesis, while its role in gastric cancer remains unclear. Methods: We assessed expression and biological behavior of OPCML in gastric cancer. Results: OPCML expression was markedly reduced in tumor tissues and cancer cell lines. Decreased OPCML expression had a significant association with unfavorable tumor stage (p = 0.007) and grading (p < 0.001). Furthermore, the results revealed that OPCML was an independent prognostic factor for overall survival in gastric cancer (p = 0.002). In addition, ectopic expression of OPCML in cancer cells significantly inhibited cell viability (p < 0.01) and colony formation (p < 0.001), arrest cell cycle in G0/G1 phase and induced apoptosis, and suppressed tumor formation in nude mice. The alterations of phosphorylation status of AKT and its substrate GSK3β, up- regulation of pro-apoptotic regulators including caspase-3, caspase-9 and PARP, and up-regulation of cell cycle regulator p27, were implicated in the biological activity of OPCML in cancer cells. Conclusion: Down-regulated OPCML expression might serve as an independent predictor for unfavorable prognosis of patients, and the biological behavior supports its role as a tumor suppressor in gastric cancer.
    [Show full text]