Matije Lucic Thesis

Total Page:16

File Type:pdf, Size:1020Kb

Matije Lucic Thesis Research Collection Doctoral Thesis The Good, the Bad and the Ugly: a three-way duel in microRNA targeting Author(s): Lucic, Matije Publication Date: 2019 Permanent Link: https://doi.org/10.3929/ethz-b-000335030 Rights / License: In Copyright - Non-Commercial Use Permitted This page was generated automatically upon download from the ETH Zurich Research Collection. For more information please consult the Terms of use. ETH Library DISS. ETH NO. 25693 The Good, the Bad and the Ugly: a three-way duel in microRNA targeting A thesis submitted to attain the degree of DOCTOR OF SCIENCES of ETH Zurich (Dr. sc. ETH Zurich) presented by MATIJE LUCIC M.Sc. Pharm. Sciences, ETH Zurich born on 09.01.1986 citizen of Muralto, Switzerland accepted on the recommendation of Prof. Dr. Jonathan Hall, examiner Prof. Dr. Constance Ciaudo, co-examiner 2019 To my loved ones. For ‘Science & Cigars’, may this be the start. ETH Zurich Matije Lucic 2 Table of contents Acknowledgements ............................................................................................................... 6 Abstract ................................................................................................................................ 7 Riassunto .............................................................................................................................. 8 1 Introduction ................................................................................................................... 9 1.1 Introduction to microRNAs (miRNAs) ..................................................................... 9 1.1.1 miRNA gene families ................................................................................... 10 1.1.2 miRNA nomenclature ................................................................................... 10 1.1.3 miRNA guide and passenger strands ........................................................... 11 1.1.4 miRBase: the miRNA sequence repository .................................................. 12 1.2 miRNA biogenesis ............................................................................................... 14 1.3 Strand selection and RISC assembly ................................................................... 15 1.4 miRNA-mediated gene silencing .......................................................................... 16 1.4.1 Argonaute-catalyzed slicing mechanism ...................................................... 17 1.4.2 Slicing-independent translational repression and mRNA decay ................... 18 1.5 miRNA target recognition ..................................................................................... 19 1.5.1 5′ end of the miRNA .................................................................................... 19 1.5.2 Central region of the miRNA ........................................................................ 20 1.5.3 3′ end of the miRNA .................................................................................... 21 1.5.4 Non-canonical miRNA targeting ................................................................... 22 1.5.5 Model for miRNA target recognition ............................................................. 23 1.5.6 Identifying the miRNA targetome ................................................................. 24 1.5.7 miRNA target prediction strategies ............................................................... 25 1.6 miRNA targets fight back on miRNAs .................................................................. 26 1.7 Complexity and redundancy of miRNA function ................................................... 26 1.8 Oncomirs and tumor suppressors ........................................................................ 27 1.8.1 miR-17~92 cluster ........................................................................................ 28 1.8.2 let-7 family ................................................................................................... 30 1.9 Aim of the project................................................................................................. 31 2 Results ........................................................................................................................ 32 2.1 Sequence homology between miR-17 and let-7 families ...................................... 32 2.2 Model for competitive non-canonical binding at let-7 target sites ......................... 33 2.3 Investigation of the let-7 transcriptome in HEK293T cells .................................... 34 2.3.1 Canonical repression by let-7a ..................................................................... 37 2.3.2 Canonical repression by miR-106a and miR-106b ....................................... 39 2.3.3 miR-106a seed-mutation abolishes canonical repression............................. 41 2.3.4 3′ end-mutated miR-106a retains canonical repressive activity .................... 43 2.3.5 miR-106a and miR-106b are unable to repress let-7 targets ........................ 45 2.3.6 Co-transfection of let-7a with either miR-106a or miR-106b ......................... 47 2.3.7 Co-transfection of let-7a with seed-mutated miR-106a ................................. 49 2.3.8 Co-transfection of let-7a with 3′ end-mutated miR-106a .............................. 51 2.4 Follow-up on putative let-7 targets: HMGA2 and LIN28B ..................................... 55 ETH Zurich Matije Lucic 3 2.5 let-7 competition by miR-106a-5p strand ............................................................. 58 2.6 Seed-homology sequences in human miRNAs .................................................... 61 3 Discussion ................................................................................................................... 64 4 Outlook........................................................................................................................ 67 5 Contributions ............................................................................................................... 68 6 Side projects ............................................................................................................... 69 6.1 RNAi activity of hybrid duplexes with parallel orientation ..................................... 69 6.2 Targeting miR-122 in RISC with conjugated antimiRs.......................................... 70 6.3 Antagonizing Lin28-pre-let-7 interaction with ‘looptomirs’ .................................... 71 6.4 Mono- and bis-labeling of pre-miRNAs ................................................................ 72 7 Materials and methods ................................................................................................ 73 7.1 Materials .............................................................................................................. 73 7.1.1 miRNAs and siRNAs .................................................................................... 73 7.1.2 Plasmids ...................................................................................................... 74 7.1.3 RT-qPCR primers ........................................................................................ 74 7.1.4 Antibodies .................................................................................................... 74 7.2 Methods .............................................................................................................. 75 7.2.1 Cultivation and maintenance of mammalian cell lines .................................. 75 7.2.2 Seeding of the cells...................................................................................... 75 7.2.3 Transient transfections of miRNAs and siRNAs ........................................... 75 7.2.4 RT-qPCR ..................................................................................................... 75 7.2.5 Western blot................................................................................................. 75 7.2.6 Cloning and transfections of luciferase reporter plasmids ............................ 76 7.2.7 Luciferase assay .......................................................................................... 76 7.2.8 RNA integrity and quantification ................................................................... 76 7.2.9 Library preparation ....................................................................................... 77 7.2.10 Clustering and sequencing........................................................................... 77 7.2.11 Analysis of sequencing data sets ................................................................. 77 7.2.12 Library ID ..................................................................................................... 78 7.2.13 miRNA target predictions ............................................................................. 79 7.2.14 Statistical analysis........................................................................................ 80 References ......................................................................................................................... 81 Supplementary information ................................................................................................. 90 Posters ............................................................................................................................. 104 Curriculum vitae ................................................................................................................ 108 ETH Zurich Matije Lucic 4 Table of figures and
Recommended publications
  • Diseasespecific and Inflammationindependent Stromal
    Full Length Arthritis & Rheumatism DOI 10.1002/art.37704 Disease-specific and inflammation-independent stromal alterations in spondyloarthritis synovitis Nataliya Yeremenko1,2, Troy Noordenbos1,2, Tineke Cantaert1,3, Melissa van Tok1,2, Marleen van de Sande1, Juan D. Cañete4, Paul P. Tak1,5*, Dominique Baeten1,2 1Department of Clinical Immunology and Rheumatology and 2Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, the Netherlands. 3Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA. 4Department of Rheumatology, Hospital Clinic de Barcelona and IDIBAPS, Spain. 5Arthrogen B.V., Amsterdam, the Netherlands. *Currently also: GlaxoSmithKline, Stevenage, U.K. Corresponding author: Dominique Baeten, MD, PhD, Department of Clinical Immunology and Rheumatology, F4-105, Academic Medical Center/University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands. E-mail: [email protected] This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process which may lead to differences between this version and the Version of Record. Please cite this article as an ‘Accepted Article’, doi: 10.1002/art.37704 © 2012 American College of Rheumatology Received: Apr 11, 2012; Revised: Jul 25, 2012; Accepted: Sep 06, 2012 Arthritis & Rheumatism Page 2 of 36 Abstract Objective: The molecular processes driving the distinct patterns of synovial inflammation and tissue remodelling in spondyloarthritis (SpA) versus rheumatoid arthritis (RA) remain largely unknown. Therefore, we aimed to identify novel and unsuspected disease- specific pathways in SpA by a systematic and unbiased synovial gene expression analysis. Methods: Differentially expressed genes were identified by pan-genomic microarray and confirmed by quantitative PCR and immunohistochemistry using synovial tissue biopsies of SpA (n=63), RA (n=28) and gout (n=9) patients.
    [Show full text]
  • Epigenetic Silencing of TGFBI Confers Resistance to Trastuzumab in Human Breast Cancer
    Palomeras et al. Breast Cancer Research (2019) 21:79 https://doi.org/10.1186/s13058-019-1160-x RESEARCH ARTICLE Open Access Epigenetic silencing of TGFBI confers resistance to trastuzumab in human breast cancer Sònia Palomeras1†, Ángel Diaz-Lagares2,3†, Gemma Viñas1,4,5, Fernando Setien2, Humberto J. Ferreira2, Glòria Oliveras1,6, Ana B. Crujeiras7,8, Alejandro Hernández4, David H. Lum9, Alana L. Welm9, Manel Esteller2,10,11,12,13* and Teresa Puig1* Abstract Background: Acquired resistance to trastuzumab is a major clinical problem in the treatment of HER2-positive (HER2+) breast cancer patients. The selection of trastuzumab-resistant patients is a great challenge of precision oncology. The aim of this study was to identify novel epigenetic biomarkers associated to trastuzumab resistance in HER2+ BC patients. Methods: We performed a genome-wide DNA methylation (450K array) and a transcriptomic analysis (RNA-Seq) comparing trastuzumab-sensitive (SK) and trastuzumab-resistant (SKTR) HER2+ human breast cancer cell models. The methylation and expression levels of candidate genes were validated by bisulfite pyrosequencing and qRT-PCR, respectively. Functional assays were conducted in the SK and SKTR models by gene silencing and overexpression. Methylation analysis in 24 HER2+ human BC samples with complete response or non-response to trastuzumab- based treatment was conducted by bisulfite pyrosequencing. Results: Epigenomic and transcriptomic analysis revealed the consistent hypermethylation and downregulation of TGFBI, CXCL2, and SLC38A1 genes in association with trastuzumab resistance. The DNA methylation and expression levels of these genes were validated in both sensitive and resistant models analyzed. Of the genes, TGFBI presented the highest hypermethylation-associated silencing both at the transcriptional and protein level.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • 0.5) in Stat3∆/∆ Compared with Stat3flox/Flox
    Supplemental Table 2 Genes down-regulated (<0.5) in Stat3∆/∆ compared with Stat3flox/flox Probe ID Gene Symbol Gene Description Entrez gene ID 1460599_at Ermp1 endoplasmic reticulum metallopeptidase 1 226090 1460463_at H60c histocompatibility 60c 670558 1460431_at Gcnt1 glucosaminyl (N-acetyl) transferase 1, core 2 14537 1459979_x_at Zfp68 zinc finger protein 68 24135 1459747_at --- --- --- 1459608_at --- --- --- 1459168_at --- --- --- 1458718_at --- --- --- 1458618_at --- --- --- 1458466_at Ctsa cathepsin A 19025 1458345_s_at Colec11 collectin sub-family member 11 71693 1458046_at --- --- --- 1457769_at H60a histocompatibility 60a 15101 1457680_a_at Tmem69 transmembrane protein 69 230657 1457644_s_at Cxcl1 chemokine (C-X-C motif) ligand 1 14825 1457639_at Atp6v1h ATPase, H+ transporting, lysosomal V1 subunit H 108664 1457260_at 5730409E04Rik RIKEN cDNA 5730409E04Rik gene 230757 1457070_at --- --- --- 1456893_at --- --- --- 1456823_at Gm70 predicted gene 70 210762 1456671_at Tbrg3 transforming growth factor beta regulated gene 3 21378 1456211_at Nlrp10 NLR family, pyrin domain containing 10 244202 1455881_at Ier5l immediate early response 5-like 72500 1455576_at Rinl Ras and Rab interactor-like 320435 1455304_at Unc13c unc-13 homolog C (C. elegans) 208898 1455241_at BC037703 cDNA sequence BC037703 242125 1454866_s_at Clic6 chloride intracellular channel 6 209195 1453906_at Med13l mediator complex subunit 13-like 76199 1453522_at 6530401N04Rik RIKEN cDNA 6530401N04 gene 328092 1453354_at Gm11602 predicted gene 11602 100380944 1453234_at
    [Show full text]
  • Investigation of Candidate Genes and Mechanisms Underlying Obesity
    Prashanth et al. BMC Endocrine Disorders (2021) 21:80 https://doi.org/10.1186/s12902-021-00718-5 RESEARCH ARTICLE Open Access Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules G. Prashanth1 , Basavaraj Vastrad2 , Anandkumar Tengli3 , Chanabasayya Vastrad4* and Iranna Kotturshetti5 Abstract Background: Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. Methods: To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. Results: A total of 820 DEGs were identified between
    [Show full text]
  • Long, Noncoding RNA Dysregulation in Glioblastoma
    cancers Review Long, Noncoding RNA Dysregulation in Glioblastoma Patrick A. DeSouza 1,2 , Xuan Qu 1, Hao Chen 1,3, Bhuvic Patel 1 , Christopher A. Maher 2,4,5,6 and Albert H. Kim 1,6,* 1 Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; [email protected] (P.A.D.); [email protected] (X.Q.); [email protected] (H.C.); [email protected] (B.P.) 2 Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; [email protected] 3 Department of Neuroscience, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA 4 Department of Biomedical Engineering, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA 5 McDonnell Genome Institute, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA 6 Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA * Correspondence: [email protected] Simple Summary: Developing effective therapies for glioblastoma (GBM), the most common primary brain cancer, remains challenging due to the heterogeneity within tumors and therapeutic resistance that drives recurrence. Noncoding RNAs are transcribed from a large proportion of the genome and remain largely unexplored in their contribution to the evolution of GBM tumors. Here, we will review the general mechanisms of long, noncoding RNAs and the current knowledge of how these impact heterogeneity and therapeutic resistance in GBM. A better understanding of the molecular drivers required for these aggressive tumors is necessary to improve the management and outcomes Citation: DeSouza, P.A.; Qu, X.; of this challenging disease.
    [Show full text]
  • Product Description SALSA MLPA Probemix P476-A1 ZNRF3
    MRC-Holland ® Product Description version A1-02; Issued 25 September 2018 MLPA Product Description SALSA ® MLPA ® Probemix P476-A1 ZNRF3 To be used with the MLPA General Protocol. Version A1. New Product. Catalogue numbers: • P476-025R: SALSA MLPA Probemix P476 ZNRF3, 25 reactions. • P476-050R: SALSA MLPA Probemix P476 ZNRF3, 50 reactions. • P476-100R: SALSA MLPA Probemix P476 ZNRF3, 100 reactions. To be used in combination with a SALSA MLPA reagent kit, available for various number of reactions. MLPA reagent kits are either provided with FAM or Cy5.0 dye-labelled PCR primer, suitable for Applied Biosystems and Beckman capillary sequencers, respectively (see www.mlpa.com ). Certificate of Analysis: Information regarding storage conditions, quality tests, and a sample electropherogram from the current sales lot is available at www.mlpa.com . Precautions and warnings: For professional use only. Always consult the most recent product description AND the MLPA General Protocol before use: www.mlpa.com . It is the responsibility of the user to be aware of the latest scientific knowledge of the application before drawing any conclusions from findings generated with this product. General information: The SALSA MLPA Probemix P476 ZNRF3 is a research use only (RUO) assay for the detection of deletions and duplications in the ZNRF3 gene. ZNRF3 is an E3 ubiquitin-protein ligase that acts as a negative feedback regulator of Wnt signalling (Hao et al. 2012). Three independent studies show homozygous deletions of the ZNRF3 gene in 10 to 16% of adrenocortical carcinoma cases (Assié et al. 2014; Juhlin et al. 2015; Zheng et al. 2016). Moreover, in 51% of microsatellite stable colorectal cancers deletion events at the ZNRF3 locus are detected (Bond et al.
    [Show full text]
  • Identification of Active Signaling Pathways by Integrating Gene Expression and Protein Interaction Data Md Humayun Kabir1,2,3, Ralph Patrick2,4,5, Joshua W
    Kabir et al. BMC Systems Biology 2018, 12(Suppl 9):120 https://doi.org/10.1186/s12918-018-0655-x RESEARCH Open Access Identification of active signaling pathways by integrating gene expression and protein interaction data Md Humayun Kabir1,2,3, Ralph Patrick2,4,5, Joshua W. K. Ho2,4,6* and Michael D. O’Connor1,7* From 29th International Conference on Genome Informatics Yunnan, China. 3-5 December 2018 Abstract Background: Signaling pathways are the key biological mechanisms that transduce extracellular signals to affect transcription factor mediated gene regulation within cells. A number of computational methods have been developed to identify the topological structure of a specific signaling pathway using protein-protein interaction data, but they are not designed for identifying active signaling pathways in an unbiased manner. On the other hand, there are statistical methods based on gene sets or pathway data that can prioritize likely active signaling pathways, but they do not make full use of active pathway structure that link receptor, kinases and downstream transcription factors. Results: Here, we present a method to simultaneously predict the set of active signaling pathways, together with their pathway structure, by integrating protein-protein interaction network and gene expression data. We evaluated the capacity for our method to predict active signaling pathways for dental epithelial cells, ocular lens epithelial cells, human pluripotent stem cell-derived lens epithelial cells, and lens fiber cells. This analysis showed our approach could identify all the known active pathways that are associated with tooth formation and lens development. Conclusions: The results suggest that SPAGI can be a useful approach to identify the potential active signaling pathways given a gene expression profile.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Familial Cortical Myoclonus Caused by Mutation in NOL3 by Jonathan Foster Rnsseil DISSERTATION Submitted in Partial Satisfaction
    Familial Cortical Myoclonus Caused by Mutation in NOL3 by Jonathan Foster Rnsseil DISSERTATION Submitted in partial satisfaction of the requirements for the degree of DOCTOR OF PHILOSOPHY in Biomedical Sciences in the Copyright 2013 by Jonathan Foster Russell ii I dedicate this dissertation to Mom and Dad, for their adamantine love and support iii No man has earned the right to intellectual ambition until he has learned to lay his course by a star which he has never seen—to dig by the divining rod for springs which he may never reach. In saying this, I point to that which will make your study heroic. For I say to you in all sadness of conviction, that to think great thoughts you must be heroes as well as idealists. Only when you have worked alone – when you have felt around you a black gulf of solitude more isolating than that which surrounds the dying man, and in hope and in despair have trusted to your own unshaken will – then only will you have achieved. Thus only can you gain the secret isolated joy of the thinker, who knows that, a hundred years after he is dead and forgotten, men who never heard of him will be moving to the measure of his thought—the subtile rapture of a postponed power, which the world knows not because it has no external trappings, but which to his prophetic vision is more real than that which commands an army. -Oliver Wendell Holmes, Jr. iv ACKNOWLEDGMENTS I am humbled by the efforts of many, many others who were essential for this work.
    [Show full text]
  • Kremen Is Beyond a Subsidiary Co-Receptor of Wnt Signaling: an in Silico Validation
    Turkish Journal of Biology Turk J Biol (2015) 39: 501-510 http://journals.tubitak.gov.tr/biology/ © TÜBİTAK Research Article doi:10.3906/biy-1409-1 Kremen is beyond a subsidiary co-receptor of Wnt signaling: an in silico validation 1 2, 3 Hemn MOHAMMADPOUR , Saeed KHALILI *, Zahra Sadat HASHEMI 1 Department of Medical Immunology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran 2 Department of Medical Biotechnology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran 3 Department of Medical Biotechnology, School of Advanced Medical Technologies, Tehran University of Medical Science, Tehran, Iran Received: 01.09.2014 Accepted/Published Online: 12.02.2015 Printed: 15.06.2015 Abstract: Kremen-1 is a co-receptor of Wnt signaling, interacting with the Dkk-3 protein, which is an antagonist of the Wnt/β catenin pathway. In the present study we attempted to shed some light on the possible orientations of Kremen/Dkk-3 interactions, explaining the mechanisms of Kremen and Dkk-3 functions. Employing state-of-the-art software, a Kremen model was built and subsequently refined. The quality of the final model was evaluated using RMSD calculations. Ultimately, we used the Kremen model for docking analysis between Kremen and Dkk-3 molecules. A model built by the Robetta server showed the best quality scores. Near native coordination of the final model was verified getting < 2 Å RMSD values between our model and an experimentally resolved structure. Docking analysis indicates that one low energy orientation for Kremen/Dkk-3 involves all extracellular domains of Kremen, while another only involves the CUB and WSC domains.
    [Show full text]
  • Meta-Analysis Identifies 13 New Loci Associated with Waist-Hip Ratio And
    ARTICLES Meta-analysis identifies 13 new loci associated with ­ waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution Waist-hip ratio (WHR) is a measure of body fat distribution and a predictor of metabolic consequences independent of overall adiposity. WHR is heritable, but few genetic variants influencing this trait have been identified. We conducted a meta-analysis of 32 genome-wide association studies for WHR adjusted for body mass index (comprising up to 77,167 participants), following up 16 loci in an additional 29 studies (comprising up to 113,636 subjects). We identified 13 new loci in or near RSPO3, VEGFA, TBX15-WARS2, NFE2L3, GRB14, DNM3-PIGC, ITPR2-SSPN, LY86, HOXC13, ADAMTS9, ZNRF3-KREMEN1, NISCH-STAB1 and CPEB4 (P = 1.9 × 10−9 to P = 1.8 × 10−40) and the known signal at LYPLAL1. Seven of these loci exhibited marked sexual dimorphism, all with a stronger effect on WHR in women than men (P for sex difference = 1.9 × 10−3 to P = 1.2 × 10−13). These findings provide evidence for multiple loci that modulate body fat distribution independent of overall adiposity and reveal strong gene-by-sex interactions. Central obesity and body fat distribution, as measured by waist ­discovery stage, up to 2,850,269 imputed and genotyped SNPs ­circumference and WHR, are associated with individual risk of type were examined in 32 GWAS comprising up to 77,167 participants 2 diabetes (T2D)1,2 and coronary heart disease3 and with mortality informative for anthropometric measures of body fat distribution. from all causes4.
    [Show full text]