American Society of Human Genetics 67Th Annual Meeting October 17–21, 2017 in Orlando, Florida

Total Page:16

File Type:pdf, Size:1020Kb

American Society of Human Genetics 67Th Annual Meeting October 17–21, 2017 in Orlando, Florida American Society of Human Genetics 67th Annual Meeting October 17–21, 2017 in Orlando, Florida PLATFORM ABSTRACTS Tuesday, October 17, 5:30-7:00 pm: Abstract #'s 4 Featured Plenary Abstract Session I South Hall B #1-#4 Wednesday, October 18, 9:00-10:30 am, Concurrent Platform Session A: 6 Genetics of Vascular, Valvular, and Syndromic Disorders Room 220B #5-#10 7 Modeling Mega-cohorts: Insights & Innovation Room 220F #11-#16 8 Genetics and Epigenetics in Mental Illness Room 230C #17-#22 9 Genome Structure and Function: The Contribution of Mutations to Human Room 230G #23-#28 Genetic Diversity, Disease, and Evolution 10 Disease Gene Discovery Strategies Room 310A #29-#34 11 Therapeutic Advances in Mendelian Disease Room 310C #35-#40 12 Detection and Impact of Mosaicism in Human Disease Room 320 #41-#46 13 Pleiotropism and Penetrance in Cancer-causing Genes Room 330A #47-#52 14 Landscape of Cancer: Bioinformatic Analyses Room 330C #53-#58 Wednesday, October 18, 11:00 am-12:30 pm, Concurrent Platform Session B: 15 Screening Cancer Cohorts for Novel Germline Cancer Genes Room 220B #59-#64 16 Cancer Genetic Testing: Approaches, Barriers, and Psychosocial Impact Room 220F #65-#70 17 Advances in Association Analysis Room 230C #71-#76 18 Strategies for Variant Interpretation Room 230G #77-#82 19 From Association to Function for Cardiometabolic Traits Room 310A #83-#88 20 Reproductive Genetics: Detection, Treatment, and Natural History of Errors Room 310C #89-#94 21 Leveraging Human Knockouts to Understand Biology Room 320 #95-#100 22 Detection and Interpretation of Structural Variation Room 330A #101-#106 23 Neurodevelopmental Disorders: Causes and Mechanisms Room 330C #107-#112 Thursday, October 19, 9:00-10:30 am, Concurrent Platform Session C: 29 Gene Discovery in Skeletal Phenotypes Room 220B #113-#118 Repeats and Rearrangements: New Methods, Genes, and Mechanisms in 30 Room 220F #119-#124 Neurological Disease 31 Secondary and Incidental Findings from WES/WGS Room 230C #125-#130 32 Computational Methods for Causal Inference in Complex Traits Room 230G #131-#136 33 Microbiome, Variation, and Disease Room 310A #137-#142 34 Genetic Architecture of Neurological Traits Room 310C #143-#148 35 High Throughput Functional Analysis of Enhancers and Variants Room 320 #149-#154 36 Genetic Architecture of Rare Variants Across Diseases Room 330A #155-#160 37 Non-coding Variation and Epigenetic Effects in Cancer Room 330C #161-#166 Thursday, October 19, 11:00 am-12:30 pm, Concurrent Platform Session D: 38 Neuromuscular Disease Room 220B #167-#172 39 Advances in the Genetics of Autoimmune Disease Room 220F #173-#178 40 Defining High Risk in Cancer Room 230C #179-#184 41 Natural Selection on Human Phenotypes Room 230G #185-#190 42 Consumers and Health Care Providers: Perspectives of Genetic Technology Room 310A #191-#196 43 Gene Discovery and Functional Models of Intellectual Disability Room 310C #197-#202 44 Polygenic Risk Scores and Genetic Correlation in Complex Disease Room 320 #203-#208 45 Single Cell Omics Technologies Room 330A #209-#214 46 Sequencing in Neonatal and Pediatric Disorders Room 330C #215-#220 Friday, October 20, 9:00-10:00 am, Concurrent Platform Session E: 56 Genomic Testing: A Focus on Results Room 220B #221-#224 57 Exploring the Impact of Archaic Ancestry Room 220F #225-#228 58 Data Sharing to Improve Genomic Variant Interpretation Room 230C #229-#232 59 Congenital and Pediatric Heart Diseases Room 230G #233-#236 60 Transcriptomics in Complex Neurological/Neuropsychiatric Disease Room 310A #237-#240 61 Context Matters: Genes, Environment, and Sex (Part 1) Room 310C #241-#244 62 New Paradigms for Regulatory Variant Contribution to Disease Risk Room 320 #245-#248 63 Gene Expression Studies of T2D Room 330A #249-#252 64 Measuring Effects of Genetic Variants with High-Throughput Assays Room 330C #253-#256 Friday, October 20, 10:15-11:15 am, Concurrent Platform Session F: 65 DNA Methylation Room 220B #257-#260 66 Splicing in Complex Traits Room 220F #261-#264 67 Ocular Development and Disease Room 230C #265-#268 68 Blood Omics in Large Cohorts Room 230G #269-#272 69 Genetics of Addictive Behaviors Room 310A #273-#276 70 Context Matters: Genes, Environment, and Sex (Part 2) Room 310C #277-#280 71 Autism Room 320 #281-#284 72 Clinical Genomics in Cancer Room 330A #285-#288 73 Transcriptomic Analysis of Genetic Variation and Disease Room 330C #289-#292 Friday, October 20, 5:30-7:00 pm: 87 Featured Plenary Abstract Session II South Hall B #293-#296 Saturday, October 21, 8:30-9:30 am, Concurrent Platform Session G: 88 Functional Analyses of Cancer Genes Room 220B #297-#300 89 Modelling Candidate Disease Variants in Cellular and Animal Models Room 220F #301-#304 90 Cerebral Palsy and Epilepsy Room 230C #305-#308 91 Diverse Approaches to the Genetics of T2D Room 230G #309-#312 92 Population-based Diagnostic Sequencing Room 310A #313-#316 93 Identification and Function of Enhancers Room 310C #317-#320 94 Genetic Associations for Behavioral Phenotypes Room 320 #321-#324 95 Host-pathogen Interactions in the Genetics of the Immune System Room 330A #325-#328 96 Investigating the Role of Non-coding Variants in Disease Room 330C #329-#332 Saturday, October 21, 9:45-10:45 am, Concurrent Platform Session H: 97 Transcriptome-wide Association Studies Room 220B #333-#336 98 Improved Interpretation of Missense Variants Room 220F #337-#340 99 Big Data Approaches in Support of Population Studies Room 230C #341-#344 100 Novel Genetic and Environmental Contributions to Cancer Risk Room 230G #345-#348 101 Neurological Disorders: Chromatin in the Spotlight Room 310A #349-#352 102 Enhancers and Human Disease Room 310C #353-#356 103 The Genetics of Obesity Room 320 #357-#360 104 Advancing Drug Discovery by Genetic Analysis in Large Cohorts Room 330A #361-#364 105 Regulation of Gene Expression in Metabolic and Vascular Tissues Room 330C #365-#368 Saturday, October 21, 11:00 am-12:30 pm: 106 Featured Plenary Abstract Session III South Hall B #369-#372 Copyright © 2017 The American Society of Human Genetics. All rights reserved. ASHG 2017 Abstracts 1 1 2 Novel loci associated with skin pigmentation identified in African popu- An atlas of 8,342 mosaic structural variants reveals genetic drivers of lations. N. Crawford1, D. Kelly1,2, M. Hansen1, M. Holsbach Beltrame1, S. Fan1, clonal hematopoiesis. P. Loh1,2, G. Genovese2,3,4, R.E. Handsaker2,3,4, H.K. S. Bowman3,4, E. Jewett5,6, A. Ranciaro1, S. Thompson1, S. Pfeifer7, J. Jensen7, Finucane1,5, Y.A. Reshef6, P. Palamara1,2, B.M. Birmann7, S.F. Bakhoum8,9, S. Wata Mpoloka9, G. Mokone10, T. Nyambo11, D. Wolde Meskel12, G. Belay12, S.A. McCarroll2,3,4, A.L. Price1,2,10. 1) Department of Epidemiology, Harvard H. Rothschild13, Y. Zhou14,15, M. Kovacs16, M. Xu16, E. Oceana20, Y. Song5,6,21,22,23, T.H. Chan School of Public Health, Boston, MA; 2) Program in Medical and 24 16 3,4 17 17 18,19 E. Eskin , K. Brown , M. Marks , S. Loftus , W. Pavan , M. Yeager , S. Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA; 3) 24 1,25 Chanock , S. Tishkoff . 1) Department of Genetics, Perelman School of Med- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, icine, University of Pennsylvania, Philadelphia, PA 19104, USA; 2) Genomics Cambridge, MA; 4) Department of Genetics, Harvard Medical School, Boston, and Computational Biology Graduate Program, University of Pennsylvania, MA; 5) Department of Mathematics, Massachusetts Institute of Technology, Philadelphia, PA, 19104, USA; 3) Department of Pathology & Laboratory Med- Cambridge, MA; 6) Department of Computer Science, Harvard University, icine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA Cambridge, MA; 7) Channing Division of Network Medicine, Department 19104; 4) Department of Pathology & Laboratory Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Phila- of Medicine, Brigham and Women’s Hospital and Harvard Medical School, delphia, PA 19104; 5) Department of EECS, University of California, Berkeley, Boston, MA; 8) Department of Radiation Oncology, Memorial Sloan Kettering CA, 94704; 6) Department of Statistics, University of California, Berkeley, CA, Cancer Center, New York, NY; 9) Sandra and Edward Meyer Cancer Center, 94704; 7) School of Life Sciences, Arizona State University, Tempe, AZ 85287; Weill Cornell Medicine, New York, NY; 10) Department of Biostatistics, Harvard 8) Department of Biology, Howard University, Washington D.C; 9) Department T.H. Chan School of Public Health, Boston, MA. of Biological Sciences, University of Botswana, Gaborone Botswana; 10) Clonal expansions of blood cells harboring somatic mutations are often Dept. of Biomedical Sciences, University of Botswana School of Medicine, observed in elderly individuals not known to have cancer (Forsberg et al. 2017 Gaborone, Botswana; 11) Department of Biochemistry, Muhimbili University Nat Rev Genet). The somatic mutations observed in clonal expansions are of Health and Allied Sciences, Dar es Salaam, Tanzania; 12) Department enriched at genes commonly mutated in cancer; accordingly, detectable clonal of Biology, Addis Ababa University, Addis Ababa, Ethiopia; 13) Stem Cell mosaicism is known to confer >10x increased risk of future hematological Program, Division of Hematology/Oncology, Pediatric Hematology Program, malignancy. However, the mechanisms that shape most clonal expansions in Boston Children's Hospital and Dana Farber Cancer Institute, Harvard Medical healthy individuals remain unknown, and the effects of specific somatic events School, Boston, MA 02115, USA; 14) Harvard Stem Cell Institute, Harvard on incident cancers have been unresolved by previous studies of up to ~1,000 University, Cambridge, MA 02138, USA; 15) Stem Cell Program, Division of detected mosaic events. Here we describe insights from an analysis of 8,342 Hematology/Oncology, Pediatric Hematology Program, Boston Children's mosaic structural variants (SVs) which we ascertained in genotyping intensity Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, data from 151,202 UK Biobank participants.
Recommended publications
  • Cytogenomic SNP Microarray - Fetal ARUP Test Code 2002366 Maternal Contamination Study Fetal Spec Fetal Cells
    Patient Report |FINAL Client: Example Client ABC123 Patient: Patient, Example 123 Test Drive Salt Lake City, UT 84108 DOB 2/13/1987 UNITED STATES Gender: Female Patient Identifiers: 01234567890ABCD, 012345 Physician: Doctor, Example Visit Number (FIN): 01234567890ABCD Collection Date: 00/00/0000 00:00 Cytogenomic SNP Microarray - Fetal ARUP test code 2002366 Maternal Contamination Study Fetal Spec Fetal Cells Single fetal genotype present; no maternal cells present. Fetal and maternal samples were tested using STR markers to rule out maternal cell contamination. This result has been reviewed and approved by Maternal Specimen Yes Cytogenomic SNP Microarray - Fetal Abnormal * (Ref Interval: Normal) Test Performed: Cytogenomic SNP Microarray- Fetal (ARRAY FE) Specimen Type: Direct (uncultured) villi Indication for Testing: Patient with 46,XX,t(4;13)(p16.3;q12) (Quest: EN935475D) ----------------------------------------------------------------- ----- RESULT SUMMARY Abnormal Microarray Result (Male) Unbalanced Translocation Involving Chromosomes 4 and 13 Classification: Pathogenic 4p Terminal Deletion (Wolf-Hirschhorn syndrome) Copy number change: 4p16.3p16.2 loss Size: 5.1 Mb 13q Proximal Region Deletion Copy number change: 13q11q12.12 loss Size: 6.1 Mb ----------------------------------------------------------------- ----- RESULT DESCRIPTION This analysis showed a terminal deletion (1 copy present) involving chromosome 4 within 4p16.3p16.2 and a proximal interstitial deletion (1 copy present) involving chromosome 13 within 13q11q12.12. This
    [Show full text]
  • Synergistic Genetic Interactions Between Pkhd1 and Pkd1 Result in an ARPKD-Like Phenotype in Murine Models
    BASIC RESEARCH www.jasn.org Synergistic Genetic Interactions between Pkhd1 and Pkd1 Result in an ARPKD-Like Phenotype in Murine Models Rory J. Olson,1 Katharina Hopp ,2 Harrison Wells,3 Jessica M. Smith,3 Jessica Furtado,1,4 Megan M. Constans,3 Diana L. Escobar,3 Aron M. Geurts,5 Vicente E. Torres,3 and Peter C. Harris 1,3 Due to the number of contributing authors, the affiliations are listed at the end of this article. ABSTRACT Background Autosomal recessive polycystic kidney disease (ARPKD) and autosomal dominant polycystic kidney disease (ADPKD) are genetically distinct, with ADPKD usually caused by the genes PKD1 or PKD2 (encoding polycystin-1 and polycystin-2, respectively) and ARPKD caused by PKHD1 (encoding fibrocys- tin/polyductin [FPC]). Primary cilia have been considered central to PKD pathogenesis due to protein localization and common cystic phenotypes in syndromic ciliopathies, but their relevance is questioned in the simple PKDs. ARPKD’s mild phenotype in murine models versus in humans has hampered investi- gating its pathogenesis. Methods To study the interaction between Pkhd1 and Pkd1, including dosage effects on the phenotype, we generated digenic mouse and rat models and characterized and compared digenic, monogenic, and wild-type phenotypes. Results The genetic interaction was synergistic in both species, with digenic animals exhibiting pheno- types of rapidly progressive PKD and early lethality resembling classic ARPKD. Genetic interaction be- tween Pkhd1 and Pkd1 depended on dosage in the digenic murine models, with no significant enhancement of the monogenic phenotype until a threshold of reduced expression at the second locus was breached.
    [Show full text]
  • Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin
    cells Review Modes of Interaction of KMT2 Histone H3 Lysine 4 Methyltransferase/COMPASS Complexes with Chromatin Agnieszka Bochy ´nska,Juliane Lüscher-Firzlaff and Bernhard Lüscher * ID Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany; [email protected] (A.B.); jluescher-fi[email protected] (J.L.-F.) * Correspondence: [email protected]; Tel.: +49-241-8088850; Fax: +49-241-8082427 Received: 18 January 2018; Accepted: 27 February 2018; Published: 2 March 2018 Abstract: Regulation of gene expression is achieved by sequence-specific transcriptional regulators, which convey the information that is contained in the sequence of DNA into RNA polymerase activity. This is achieved by the recruitment of transcriptional co-factors. One of the consequences of co-factor recruitment is the control of specific properties of nucleosomes, the basic units of chromatin, and their protein components, the core histones. The main principles are to regulate the position and the characteristics of nucleosomes. The latter includes modulating the composition of core histones and their variants that are integrated into nucleosomes, and the post-translational modification of these histones referred to as histone marks. One of these marks is the methylation of lysine 4 of the core histone H3 (H3K4). While mono-methylation of H3K4 (H3K4me1) is located preferentially at active enhancers, tri-methylation (H3K4me3) is a mark found at open and potentially active promoters. Thus, H3K4 methylation is typically associated with gene transcription. The class 2 lysine methyltransferases (KMTs) are the main enzymes that methylate H3K4. KMT2 enzymes function in complexes that contain a necessary core complex composed of WDR5, RBBP5, ASH2L, and DPY30, the so-called WRAD complex.
    [Show full text]
  • 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]
  • Genome-Wide Parent-Of-Origin DNA Methylation Analysis Reveals The
    Downloaded from genome.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press Research Genome-wide parent-of-origin DNA methylation analysis reveals the intricacies of human imprinting and suggests a germline methylation-independent mechanism of establishment Franck Court,1,15 Chiharu Tayama,2,15 Valeria Romanelli,1,15 Alex Martin-Trujillo,1,15 Isabel Iglesias-Platas,3 Kohji Okamura,4 Naoko Sugahara,2 Carlos Simo´n,5 Harry Moore,6 Julie V. Harness,7 Hans Keirstead,7 Jose Vicente Sanchez-Mut,8 Eisuke Kaneki,9 Pablo Lapunzina,10 Hidenobu Soejima,11 Norio Wake,9 Manel Esteller,8,12,13 Tsutomu Ogata,14 Kenichiro Hata,2 Kazuhiko Nakabayashi,2,16,17 and David Monk1,16,17 1–14[Author affiliations appear at the end of the paper.] Differential methylation between the two alleles of a gene has been observed in imprinted regions, where the methylation of one allele occurs on a parent-of-origin basis, the inactive X-chromosome in females, and at those loci whose methylation is driven by genetic variants. We have extensively characterized imprinted methylation in a substantial range of normal human tissues, reciprocal genome-wide uniparental disomies, and hydatidiform moles, using a combination of whole- genome bisulfite sequencing and high-density methylation microarrays. This approach allowed us to define methylation profiles at known imprinted domains at base-pair resolution, as well as to identify 21 novel loci harboring parent-of-origin methylation, 15 of which are restricted to the placenta. We observe that the extent of imprinted differentially methylated regions (DMRs) is extremely similar between tissues, with the exception of the placenta.
    [Show full text]
  • KAT6A Amplifications Are Associated with Shorter Progression-Free Survival and Overall Survival in Patients with Endometrial Serous Carcinoma
    PLOS ONE RESEARCH ARTICLE KAT6A amplifications are associated with shorter progression-free survival and overall survival in patients with endometrial serous carcinoma 1 2 2 2 2 Ozlen Saglam , Zhenya Tang , Guilin Tang , L. Jeffrey Medeiros , Gokce A. TorunerID * 1 Department of Surgical Pathology, Moffitt Cancer Center, Tampa, Florida, United States of America, 2 Department of Hematopathology, Section of Clinical Cytogenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America * [email protected] a1111111111 a1111111111 Abstract a1111111111 a1111111111 Somatic copy number alterations (CNA) are common in endometrial serous carcinoma a1111111111 (ESC). We used the Tumor Cancer Genome Atlas Pan Cancer dataset (TCGA Pan Can) to explore the impact of somatic CNA and gene expression levels (mRNA) of cancer-related genes in ESC. Results were correlated with clinico-pathologic parameters such as age of onset, disease stage, progression-free survival (PFS) and overall survival (OS) (n = 108). OPEN ACCESS 1,449 genes with recurrent somatic CNA were identified, observed in 10% or more tumor Citation: Saglam O, Tang Z, Tang G, Medeiros LJ, samples. Somatic CNA and mRNA expression levels were highly correlated (r> = 0.6) for Toruner GA (2020) KAT6A amplifications are 383 genes. Among these, 45 genes were classified in the Tier 1 category of Cancer associated with shorter progression-free survival and overall survival in patients with endometrial Genome Census-Catalogue of Somatic Mutations in Cancer. Eighteen of 45 Tier 1 genes serous carcinoma. PLoS ONE 15(9): e0238477. had highly correlated somatic CNA and mRNA expression levels including ARNT, PIK3CA, https://doi.org/10.1371/journal.pone.0238477 TBLXR1, ASXL1, EIF4A2, HOOK3, IKBKB, KAT6A, TCEA1, KAT6B, ERBB2, BRD4, Editor: JesuÂs Maria Paramio, CIEMAT, SPAIN KEAP1, PRKACA, DNM2, SMARCA4, AKT2, SS18L1.
    [Show full text]
  • Evaluation of GKN1 and GKN2 Gene Expression As a Biomarker of Gastric Cancer
    Archive of SID Gastroenterology and Hepatology From Bed to Bench. BRIEF REPORT ©2018 RIGLD, Research Institute for Gastroenterology and Liver Diseases Evaluation of GKN1 and GKN2 gene expression as a biomarker of gastric cancer Fatemeh Dokhaee1, Sogol Mazhari2, Mohammad Galehdari1, Ayad Bahadori Monfared3, Kaveh Baghaei4 1 Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran 2 Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Science, Tehran, Iran 3 Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran 4 Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran ABSTRACT Aim: The aim of this study was to investigate the expression of GKN1 and GKN2 genes as probable biomarkers for gastric cancer. Background: Gastric cancer is a multifactorial process characterized by the uncontrolled growth and dissemination of abnormal cells. Survival rates of gastric cancer tend to be poor, a plausible explanation is a combination of a late-stage diagnosis and limited access to treatment. In this regard, finding relevant and measurable biomarkers is urgently needed. Methods: 27 samples of gastric cancer tissues were enrolled into this study, according to their pathological responses. The alteration of genes expression were evaluated by Real-Time PCR technique. Results: Our findings showed the significant reduction of Gastrokin-1 and Gastrokine-2 genes expression in the cancerous specimens in comparison with the normal tissues. (P = 0.008 and P = 0.004 respectively). Conclusion: Our findings showed the significant reduction of Gastrokin-1 and Gastrokine-2 genes expression in the cancerous specimens in comparison with the normal tissues.
    [Show full text]
  • Utx Is Required for Proper Induction of Ectoderm and Mesoderm During Differentiation of Embryonic Stem Cells
    Utx Is Required for Proper Induction of Ectoderm and Mesoderm during Differentiation of Embryonic Stem Cells Cristina Morales Torres1,2, Anne Laugesen1,2, Kristian Helin1,2,3* 1 Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark, 2 Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark, 3 The Danish Stem Cell Center, University of Copenhagen, Copenhagen, Denmark Abstract Embryonic development requires chromatin remodeling for dynamic regulation of gene expression patterns to ensure silencing of pluripotent transcription factors and activation of developmental regulators. Demethylation of H3K27me3 by the histone demethylases Utx and Jmjd3 is important for the activation of lineage choice genes in response to developmental signals. To further understand the function of Utx in pluripotency and differentiation we generated Utx knockout embryonic stem cells (ESCs). Here we show that Utx is not required for the proliferation of ESCs, however, Utx contributes to the establishment of ectoderm and mesoderm in vitro. Interestingly, this contribution is independent of the catalytic activity of Utx. Furthermore, we provide data showing that the Utx homologue, Uty, which is devoid of detectable demethylase activity, and Jmjd3 partly compensate for the loss of Utx. Taken together our results show that Utx is required for proper formation of ectoderm and mesoderm in vitro, and that Utx, similar to its C.elegans homologue, has demethylase dependent and independent functions. Citation: Morales Torres C, Laugesen A, Helin K (2013) Utx Is Required for Proper Induction of Ectoderm and Mesoderm during Differentiation of Embryonic Stem Cells. PLoS ONE 8(4): e60020. doi:10.1371/journal.pone.0060020 Editor: Qiang Wu, National University of Singapore, Singapore Received December 13, 2012; Accepted February 21, 2013; Published April 3, 2013 Copyright: ß 2013 Morales Torres et al.
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • Upregulation of E3 Ubiquitin Ligase CBLC Enhances EGFR
    Published OnlineFirst June 26, 2018; DOI: 10.1158/0008-5472.CAN-17-3858 Cancer Tumor Biology and Immunology Research Upregulation of E3 Ubiquitin Ligase CBLC Enhances EGFR Dysregulation and Signaling in Lung Adenocarcinoma Shiao-Ya Hong1, Yu-Rung Kao1, Te-Chang Lee1, and Cheng-Wen Wu1,2,3,4 Abstract CBLC (CBL proto-oncogene c) belongs to the CBL protein ubiquitinated and positively regulated aEGFR stability family, which has E3 ubiquitin ligase activity toward activated through the conjugation of polyubiquitin by K6 and K11 receptor tyrosine kinases. CBLC is frequently upregulated in linkages. This CBLC-mediated polyubiquitination promoted non–small cell lung cancer (NSCLC), yet very little is known either preferential recycling of aEGFR back to the plasma about the functions of CBLC in tumorigenesis. Here we show membrane or trafficking to the cell nucleus. IHC analyses that CBLC is an epigenetically demethylated target and its revealed a positive correlation between phospho-EGFR and expression can be upregulated in NSCLC after treatment with CBLC in lung adenocarcinoma. In summary, we demonstrate a the DNA methylation inhibitor 50-azacytidine. Depletion novel mechanism by which aEGFR escapes lysosomal degra- of CBLC significantly inhibited cell viability and clonogenicity dation in a CBLC/ubiquitin-dependent manner to sustain its in vitro and reduced tumor growth in a xenograft model. CBLC activation. Our work identifies CBLC as a potential diagnostic silencing further sensitized EGFR-mutated NSCLC cells to biomarker and also points to its utilization as a novel thera- treatment with tyrosine kinase inhibitors. Conversely, ectopic peutic target for NSCLC therapy. expression of CBLC enhanced the activation of EGFR and downstream ERK1/2 signaling after ligand stimulation by Significance: This work demonstrates the role of CBLC expres- competing with CBL for EGFR binding.
    [Show full text]
  • A Set of Regulatory Genes Co-Expressed in Embryonic Human Brain Is Implicated in Disrupted Speech Development
    Molecular Psychiatry https://doi.org/10.1038/s41380-018-0020-x ARTICLE A set of regulatory genes co-expressed in embryonic human brain is implicated in disrupted speech development 1 1 1 2 3 Else Eising ● Amaia Carrion-Castillo ● Arianna Vino ● Edythe A. Strand ● Kathy J. Jakielski ● 4,5 6 7 8 9 Thomas S. Scerri ● Michael S. Hildebrand ● Richard Webster ● Alan Ma ● Bernard Mazoyer ● 1,10 4,5 6,11 6,12 13 Clyde Francks ● Melanie Bahlo ● Ingrid E. Scheffer ● Angela T. Morgan ● Lawrence D. Shriberg ● Simon E. Fisher 1,10 Received: 22 September 2017 / Revised: 3 December 2017 / Accepted: 2 January 2018 © The Author(s) 2018. This article is published with open access Abstract Genetic investigations of people with impaired development of spoken language provide windows into key aspects of human biology. Over 15 years after FOXP2 was identified, most speech and language impairments remain unexplained at the molecular level. We sequenced whole genomes of nineteen unrelated individuals diagnosed with childhood apraxia of speech, a rare disorder enriched for causative mutations of large effect. Where DNA was available from unaffected parents, CHD3 SETD1A WDR5 fi 1234567890();,: we discovered de novo mutations, implicating genes, including , and . In other probands, we identi ed novel loss-of-function variants affecting KAT6A, SETBP1, ZFHX4, TNRC6B and MKL2, regulatory genes with links to neurodevelopment. Several of the new candidates interact with each other or with known speech-related genes. Moreover, they show significant clustering within a single co-expression module of genes highly expressed during early human brain development. This study highlights gene regulatory pathways in the developing brain that may contribute to acquisition of proficient speech.
    [Show full text]
  • Whole Genome Sequencing of Familial Non-Medullary Thyroid Cancer Identifies Germline Alterations in MAPK/ERK and PI3K/AKT Signaling Pathways
    biomolecules Article Whole Genome Sequencing of Familial Non-Medullary Thyroid Cancer Identifies Germline Alterations in MAPK/ERK and PI3K/AKT Signaling Pathways Aayushi Srivastava 1,2,3,4 , Abhishek Kumar 1,5,6 , Sara Giangiobbe 1, Elena Bonora 7, Kari Hemminki 1, Asta Försti 1,2,3 and Obul Reddy Bandapalli 1,2,3,* 1 Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany; [email protected] (A.S.); [email protected] (A.K.); [email protected] (S.G.); [email protected] (K.H.); [email protected] (A.F.) 2 Hopp Children’s Cancer Center (KiTZ), D-69120 Heidelberg, Germany 3 Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany 4 Medical Faculty, Heidelberg University, D-69120 Heidelberg, Germany 5 Institute of Bioinformatics, International Technology Park, Bangalore 560066, India 6 Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India 7 S.Orsola-Malphigi Hospital, Unit of Medical Genetics, 40138 Bologna, Italy; [email protected] * Correspondence: [email protected]; Tel.: +49-6221-42-1709 Received: 29 August 2019; Accepted: 10 October 2019; Published: 13 October 2019 Abstract: Evidence of familial inheritance in non-medullary thyroid cancer (NMTC) has accumulated over the last few decades. However, known variants account for a very small percentage of the genetic burden. Here, we focused on the identification of common pathways and networks enriched in NMTC families to better understand its pathogenesis with the final aim of identifying one novel high/moderate-penetrance germline predisposition variant segregating with the disease in each studied family.
    [Show full text]