Prenatal Diagnosis of Fetuses with Increased Nuchal Translucency By

Total Page:16

File Type:pdf, Size:1020Kb

Prenatal Diagnosis of Fetuses with Increased Nuchal Translucency By bioRxiv preprint doi: https://doi.org/10.1101/667311; this version posted June 11, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. 1 Prenatal Diagnosis of Fetuses with Increased Nuchal 2 Translucency by Genome Sequencing Analysis 3 4 1 bioRxiv preprint doi: https://doi.org/10.1101/667311; this version posted June 11, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. 1 Prenatal Diagnosis of Fetuses with Increased Nuchal 2 Translucency by Genome Sequencing Analysis 3 Kwong Wai Choy,1,2,3# Huilin Wang,4# Mengmeng Shi,1# Jingsi Chen,5# Zhenjun Yang,1 4 Rui Zhang,4 Huanchen Yan,5 Yanfang Wang,4 Shaoyun Chen,4 Matthew Hoi Kin Chau,1 5 Ye Cao,1,6 Olivia YM Chan,1 Yvonne K Kwok,1 Yuanfang Zhu,4 Min Chen,5 Tak Yeung 6 Leung,1,2,3 Zirui Dong1,2,5* 7 1. Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, 8 Hong Kong, China 9 2. Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, 10 China 11 3. The Chinese University of Hong Kong-Baylor College of Medicine Joint Center 12 for Medical Genetics, Hong Kong, China 13 4. Department of Central Laboratory, Bao'an Maternity and Child Healthcare 14 Hospital Affiliated to Jinan University School of Medicine, Key Laboratory of Birth 15 Defects Research, Birth Defects Prevention Research and Transformation Team, 16 Shenzhen, China 17 5. Department of Obstetrics and Gynecology, Key Laboratory of Reproduction and 18 Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of 19 Guangzhou Medical University, Guangzhou, Guangdong, China 20 6. Department of Molecular and Cellular Biology, Baylor College of Medicine 21 Houston, Texas, USA 22 #These authors contributed equally. 2 bioRxiv preprint doi: https://doi.org/10.1101/667311; this version posted June 11, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. 1 *Email addresses for correspondence: [email protected] 3 bioRxiv preprint doi: https://doi.org/10.1101/667311; this version posted June 11, 2019. 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. 1 Abstract 2 Background: 3 Increased Nuchal Translucency (NT) is an important biomarker associated with increased 4 risk of fetal structural anomalies. It is known to be contributed by a wide range of genetic 5 etiologies from single nucleotide variants to those affecting millions of base-pairs. 6 Currently, prenatal diagnosis is routinely performed by karyotyping and chromosomal 7 microarray analysis (CMA), however, both of them have limited resolution. The diversity 8 of the genetic etiologies warrants an integrated assay such as genome sequencing (GS) 9 for comprehensive detection of genomic variants. Herein, we aim to evaluate the 10 feasibility of applying GS in prenatal diagnosis for the fetuses with increased NT. 11 Methods: 12 We retrospectively applied GS (>30-fold) for fetuses with increased NT (≥3.5-mm), who 13 underwent routine prenatal diagnosis. Detection of single-nucleotide variants, copy- 14 number variants and structural rearrangements was performed simultaneously and the 15 results were integrated for interpretation in accordance with the guidelines of the 16 American College of Medical Genetics and Genomics. Pathogenic or likely pathogenic 17 (P/LP) variants were selected for validation and parental confirmation, when available. 18 Results: 19 Overall, 50 fetuses were enrolled, including 34 cases with isolated increased NT and 16 20 cases with other fetal structural malformations. Routine CMA and karyotyping reported 21 eight P/LP CNVs, yielding a diagnostic rate of 16.0% (8/50). In comparison, GS 22 provided a 2-fold increase in diagnostic yield (32.0%, 16/50), including one mosaic 4 bioRxiv preprint doi: https://doi.org/10.1101/667311; this version posted June 11, 2019. 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. 1 turner syndrome, eight cases with microdeletions/microduplications and seven cases with 2 P/LP point mutations. Moreover, GS identified two cryptic insertions and two inversions. 3 Follow-up study further demonstrated the potential pathogenicity of an apparently 4 balanced insertion which disrupted an OMIM autosomal dominant disease-causing gene 5 at the inserted site. 6 Conclusions: 7 Our study demonstrates that applying GS in fetuses with increased NT can 8 comprehensively detect and delineate the various genomic variants that are causative to 9 the diseases. Importantly, prenatal diagnosis by GS doubled the diagnostic yield 10 compared with routine protocols. Given a comparable turn-around-time and less DNA 11 required, our study provides strong evidence to facilitate GS in prenatal diagnosis, 12 particularly in fetuses with increased NT. 13 14 Keywords: 15 Increased Nuchal Translucency; Genome Sequencing; Prenatal Diagnosis; Genomic 16 Variants; Structural Rearrangement 17 18 5 bioRxiv preprint doi: https://doi.org/10.1101/667311; this version posted June 11, 2019. 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. 1 Introduction 2 Detection of fetuses with increased nuchal translucency (NT) in routine first-trimester 3 ultrasound screening has been widely used as a sensitive indication for fetal chromosomal 4 abnormalities and/or fetal structural anomalies, such as congenital heart disorders or 5 neurodevelopmental anomalies detected in later gestations (Leung et al., 2011;Huang et 6 al., 2014;Socolov et al., 2017;Sinajon et al., 2019). Fetuses with increased NT and 7 structural malformations are frequently contributed by genetic abnormalities and have 8 poor prognoses. However, more than 80% of such cases do not obtain a causative result 9 with the current routine prenatal diagnostic tests (Leung et al., 2011;Huang et al., 10 2014;Yang et al., 2017;Sinajon et al., 2019), challenging genetic counseling and clinical 11 management. In addition, pathogenic copy-number variants (CNVs) only account for 0.8 12 to 5.3% of these fetuses with isolated increased NT (with/without other soft markers) 13 (Leung et al., 2011;Huang et al., 2014), and part of these cases would also have poor 14 outcomes. Therefore, a test for the comprehensive detection of disease associated 15 genomic variants including numerical disorders, structural rearrangements, CNVs and 16 point mutations in this prenatal cohort is warranted. 17 In prenatal diagnosis, quantitative fluorescent PCR (QF-PCR) is routinely conducted 18 for the detection of maternal cell contamination (MCC) and common aneuploidy [such as 19 Trisomy 21 (Choy et al., 2014;Sinajon et al., 2019)]. In addition, since 2010, 20 chromosomal microarray analysis (CMA) has been recommended as the first-tier test for 21 high-risk pregnancies in identification of microscopic or submicroscopic CNVs (Leung et 22 al., 2011;Huang et al., 2014;Sinajon et al., 2019). However, this approach is limited by its 23 resolution and it cannot detect single nucleotide variants (SNVs) and small 6 bioRxiv preprint doi: https://doi.org/10.1101/667311; this version posted June 11, 2019. 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. 1 insertions/deletions (InDels). Owing to the breakthrough of molecular technologies such 2 as next-generation sequencing and its reduction of costs over the years, whole-exome 3 sequencing (or exome sequencing, WES) has been applied for both research purposes and 4 clinical use (Drury et al., 2015;Fu et al., 2018;Leung et al., 2018;Normand et al., 5 2018;Lord et al., 2019;Petrovski et al., 2019). Emerging studies show WES has the 6 ability to provide genetic diagnoses ranging from 9.1% to 32% for the fetuses with a 7 structural anomaly (Drury et al., 2015;Fu et al., 2018;Leung et al., 2018;Normand et al., 8 2018;Lord et al., 2019;Petrovski et al., 2019), while among these cases, WES yielded 9 diagnoses in 3.2% to 21% of the fetuses with increased NT with/without structural 10 malformations (Drury et al., 2015;Lord et al., 2019;Petrovski et al., 2019). However, 11 most of these studies were conducted on prenatal cohorts after the exclusion of abnormal 12 karyotypes and/or CMA results attributed to the cost and the limited ability of WES in 13 CNV detection (Belkadi et al., 2015). These studies show the clinical utility of WES and 14 CMA in prenatal diagnosis and warrants a combination of these two approaches for each 15 case. Meanwhile, both WES and CMA are unable to detect apparently balanced structural 16 rearrangements (or structural variants, SVs), a common limitation of the current methods 17 but some of these rearrangements have been demonstrated to be disease-causing 18 (Talkowski et al., 2012a). The wide spectrum of genetic etiologies in fetuses with 19 increased NT ranging from single-base mutations to those affecting millions of base-pairs 20 and numerical disorders, warrants a holistic approach for comprehensive detection of the 21 disease-causing genetic variants. 22 Our previous studies have demonstrated the feasibility and potential diagnostic utility 23 of applying low-pass whole-genome sequencing (or genome sequencing, GS) analysis in 7 bioRxiv preprint doi: https://doi.org/10.1101/667311; this version posted June 11, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.
Recommended publications
  • Effects of Rapamycin on Social Interaction Deficits and Gene
    Kotajima-Murakami et al. Molecular Brain (2019) 12:3 https://doi.org/10.1186/s13041-018-0423-2 RESEARCH Open Access Effects of rapamycin on social interaction deficits and gene expression in mice exposed to valproic acid in utero Hiroko Kotajima-Murakami1,2, Toshiyuki Kobayashi3, Hirofumi Kashii1,4, Atsushi Sato1,5, Yoko Hagino1, Miho Tanaka1,6, Yasumasa Nishito7, Yukio Takamatsu7, Shigeo Uchino1,2 and Kazutaka Ikeda1* Abstract The mammalian target of rapamycin (mTOR) signaling pathway plays a crucial role in cell metabolism, growth, and proliferation. The overactivation of mTOR has been implicated in the pathogenesis of syndromic autism spectrum disorder (ASD), such as tuberous sclerosis complex (TSC). Treatment with the mTOR inhibitor rapamycin improved social interaction deficits in mouse models of TSC. Prenatal exposure to valproic acid (VPA) increases the incidence of ASD. Rodent pups that are exposed to VPA in utero have been used as an animal model of ASD. Activation of the mTOR signaling pathway was recently observed in rodents that were exposed to VPA in utero, and rapamycin ameliorated social interaction deficits. The present study investigated the effect of rapamycin on social interaction deficits in both adolescence and adulthood, and gene expressions in mice that were exposed to VPA in utero. We subcutaneously injected 600 mg/kg VPA in pregnant mice on gestational day 12.5 and used the pups as a model of ASD. The pups were intraperitoneally injected with rapamycin or an equal volume of vehicle once daily for 2 consecutive days. The social interaction test was conducted in the offspring after the last rapamycin administration at 5–6 weeks of ages (adolescence) or 10–11 weeks of age (adulthood).
    [Show full text]
  • Supplemental Information
    Supplemental information Dissection of the genomic structure of the miR-183/96/182 gene. Previously, we showed that the miR-183/96/182 cluster is an intergenic miRNA cluster, located in a ~60-kb interval between the genes encoding nuclear respiratory factor-1 (Nrf1) and ubiquitin-conjugating enzyme E2H (Ube2h) on mouse chr6qA3.3 (1). To start to uncover the genomic structure of the miR- 183/96/182 gene, we first studied genomic features around miR-183/96/182 in the UCSC genome browser (http://genome.UCSC.edu/), and identified two CpG islands 3.4-6.5 kb 5’ of pre-miR-183, the most 5’ miRNA of the cluster (Fig. 1A; Fig. S1 and Seq. S1). A cDNA clone, AK044220, located at 3.2-4.6 kb 5’ to pre-miR-183, encompasses the second CpG island (Fig. 1A; Fig. S1). We hypothesized that this cDNA clone was derived from 5’ exon(s) of the primary transcript of the miR-183/96/182 gene, as CpG islands are often associated with promoters (2). Supporting this hypothesis, multiple expressed sequences detected by gene-trap clones, including clone D016D06 (3, 4), were co-localized with the cDNA clone AK044220 (Fig. 1A; Fig. S1). Clone D016D06, deposited by the German GeneTrap Consortium (GGTC) (http://tikus.gsf.de) (3, 4), was derived from insertion of a retroviral construct, rFlpROSAβgeo in 129S2 ES cells (Fig. 1A and C). The rFlpROSAβgeo construct carries a promoterless reporter gene, the β−geo cassette - an in-frame fusion of the β-galactosidase and neomycin resistance (Neor) gene (5), with a splicing acceptor (SA) immediately upstream, and a polyA signal downstream of the β−geo cassette (Fig.
    [Show full text]
  • A Mode of Cell Adhesion and Migration Facilitated by CD44-Dependent Microtentacles
    A mode of cell adhesion and migration facilitated by CD44-dependent microtentacles Kayla J. Wolfa,b, Poojan Shuklab, Kelsey Springerb, Stacey Leea,b, Jason D. Coombesb,c, Caleb J. Choyd, Samuel J. Kennye,KeXue,f, and Sanjay Kumara,b,g,1 aUniversity of California, Berkeley–University of California San Francisco Graduate Program in Bioengineering, Department of Bioengineering, University of California, Berkeley, CA 94720; bDepartment of Bioengineering, University of California, Berkeley, CA, 94720; cInflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom, SE5 9NU; dDepartment of Molecular and Cell Biology, University of California, Berkeley, CA 94720; eDepartment of Chemistry, University of California, Berkeley, CA 94720; fDivision of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; and gDepartment of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720 Edited by David A. Weitz, Harvard University, Cambridge, MA, and approved March 25, 2020 (received for review August 19, 2019) The structure and mechanics of many connective tissues are grafted C6 gliomas (14). Furthermore, knockdown (KD) of dictated by a collagen-rich extracellular matrix (ECM), where col- CD44 in human GBM tumors slows tumor growth and sensitizes lagen fibers provide topological cues that direct cell migration. tumors to cytotoxic agents (15). CD44 is also a marker of glioma However, comparatively little is known about how cells navigate stem cells (GSCs) (also known as tumor-initiating cells) and the hyaluronic acid (HA)-rich, nanoporous ECM of the brain, a contributes to maintaining stemness (16). Our laboratory has problem with fundamental implications for development, inflam- shown that CD44 is necessary for adhesion and migration on mation, and tumor invasion.
    [Show full text]
  • A High Throughput, Functional Screen of Human Body Mass Index GWAS Loci Using Tissue-Specific Rnai Drosophila Melanogaster Crosses Thomas J
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 2018 A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses Thomas J. Baranski Washington University School of Medicine in St. Louis Aldi T. Kraja Washington University School of Medicine in St. Louis Jill L. Fink Washington University School of Medicine in St. Louis Mary Feitosa Washington University School of Medicine in St. Louis Petra A. Lenzini Washington University School of Medicine in St. Louis See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Baranski, Thomas J.; Kraja, Aldi T.; Fink, Jill L.; Feitosa, Mary; Lenzini, Petra A.; Borecki, Ingrid B.; Liu, Ching-Ti; Cupples, L. Adrienne; North, Kari E.; and Province, Michael A., ,"A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses." PLoS Genetics.14,4. e1007222. (2018). https://digitalcommons.wustl.edu/open_access_pubs/6820 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. Authors Thomas J. Baranski, Aldi T. Kraja, Jill L. Fink, Mary Feitosa, Petra A. Lenzini, Ingrid B. Borecki, Ching-Ti Liu, L. Adrienne Cupples, Kari E. North, and Michael A. Province This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/6820 RESEARCH ARTICLE A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses Thomas J.
    [Show full text]
  • The Akt-Mtor Pathway Drives Myelin Sheath Growth by Regulating Cap-Dependent Translation
    bioRxiv preprint doi: https://doi.org/10.1101/2021.04.12.439555; this version posted April 12, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Title Page Manuscript Title: The Akt-mTOR pathway drives myelin sheath growth by regulating cap-dependent translation Abbreviated Title: mTOR-mediated translation drives myelination Authors: Karlie N. Fedder-Semmes1,2, Bruce Appel1,3 Affiliations: 1Department of Pediatrics, Section of Developmental Biology 2Neuroscience Graduate Program 3Children’s Hospital Colorado University of Colorado Anschutz Medical Campus Aurora, Colorado, United States of America, 80045 Corresponding author: Bruce Appel, [email protected] Number of pages: 77, including references and figures Number of figures: 8 Acknowledgements: We thank the Appel lab for helpful discussions, Drs. Caleb Doll and Alexandria Hughes for constructive comments on the manuscript, Dr. Katie Yergert for experimental guidance, and Rebecca O’Rourke for help with bioinformatics analysis. This work was supported by US National Institutes of Health (NIH) grant R01 NS095670 and a gift from the Gates Frontiers Fund to B.A. K.N.F-S. was supported by NIH F31 NS115261 and NIH T32 NS099042. The University of Colorado Anschutz Medical Campus Zebrafish Core Facility was supported by NIH grant P30 NS048154. Conflict of Interest: The authors declare no competing financial interests. bioRxiv preprint doi: https://doi.org/10.1101/2021.04.12.439555; this version posted April 12, 2021.
    [Show full text]
  • A High Throughput, Functional Screen of Human Body Mass Index GWAS Loci Using Tissue-Specific Rnai Drosophila Melanogaster Crosses
    RESEARCH ARTICLE A high throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi Drosophila melanogaster crosses Thomas J. Baranski1*, Aldi T. Kraja2, Jill L. Fink1, Mary Feitosa2, Petra A. Lenzini2, Ingrid B. Borecki3, Ching-Ti Liu4, L. Adrienne Cupples4, Kari E. North5, Michael A. Province2* a1111111111 1 Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, United States of America, 2 Department of Genetics and a1111111111 Center for Genome Sciences and Systems Biology, Division of Statistical Genomics, Washington University a1111111111 School of Medicine, St. Louis, Missouri, United States of America, 3 Department of Biostatistics, University of a1111111111 Washington, Seattle, Washington, United States of America, 4 Department of Biostatistics, Boston University a1111111111 School of Public Health, Boston, Massachusetts, United States of America, 5 Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, United States of America * [email protected] (TJB); [email protected] (MAP) OPEN ACCESS Citation: Baranski TJ, Kraja AT, Fink JL, Feitosa M, Abstract Lenzini PA, Borecki IB, et al. (2018) A high Human GWAS of obesity have been successful in identifying loci associated with adiposity, throughput, functional screen of human Body Mass Index GWAS loci using tissue-specific RNAi but for the most part, these are non-coding SNPs whose function, or even whose gene of Drosophila melanogaster crosses. PLoS Genet 14 action, is unknown. To help identify the genes on which these human BMI loci may be oper- (4): e1007222. https://doi.org/10.1371/journal. ating, we conducted a high throughput screen in Drosophila melanogaster.
    [Show full text]
  • Pkcε Switches Aurora B Specificity to Exit the Abscission Checkpoint
    ARTICLE Received 4 Jan 2016 | Accepted 3 Nov 2016 | Published 22 Dec 2016 DOI: 10.1038/ncomms13853 OPEN PKCe switches Aurora B specificity to exit the abscission checkpoint Tanya Pike1, Nicola Brownlow1,w, Svend Kjaer2, Jeremy Carlton3 & Peter J. Parker1,3 The ‘NoCut’, or Aurora B abscission checkpoint can be activated if DNA is retained in the cleavage furrow after completion of anaphase. Checkpoint failure leads to incomplete abscission and a binucleate outcome. These phenotypes are also observed after loss of PKCe in transformed cell models. Here we show that PKCe directly modulates the Aurora B-dependent abscission checkpoint by phosphorylating Aurora B at S227. This phosphorylation invokes a switch in Aurora B specificity, with increased phosphorylation of a subset of target substrates, including the CPC subunit Borealin. This switch is essential for abscission checkpoint exit. Preventing the phosphorylation of Borealin leads to abscission failure, as does expression of a non-phosphorylatable Aurora B S227A mutant. Further, depletion of the ESCRT-III component and Aurora B substrate CHMP4C enables abscission, bypassing the PKCe–Aurora B exit pathway. Thus, we demonstrate that PKCe signals through Aurora B to exit the abscission checkpoint and complete cell division. 1 Protein Phosphorylation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK. 2 Protein Purification Facility, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK. 3 Division of Cancer Studies King’s College London, New Hunt’s House, Guy’s Campus, London SE1 1UL, UK. w Present address: Centre for Genomic Regulation (CRG), Dr Aiguader, 88, PRBB Building, 08003 Barcelona, Spain.
    [Show full text]
  • Clinicogenomic Analysis of FGFR2-Rearranged Cholangiocarcinoma Identifies
    Author Manuscript Published OnlineFirst on November 20, 2020; DOI: 10.1158/2159-8290.CD-20-0766 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Clinicogenomic analysis of FGFR2-rearranged cholangiocarcinoma identifies correlates of response and mechanisms of resistance to pemigatinib Ian M. Silverman,1 Antoine Hollebecque,2 Luc Friboulet,2 Sherry Owens,1 Robert C. Newton,1 Huiling Zhen,3 Luis Féliz,4 Camilla Zecchetto,5 Davide Melisi,5 and Timothy C. Burn1 1Incyte Research Institute, Wilmington, Delaware. 2Institut Gustave Roussy, Villejuif, France. 3Incyte Corporation, Wilmington, Delaware. 4Incyte Biosciences International Sàrl, Morges, Switzerland. 5Digestive Molecular Clinical Oncology Research Unit, Section of Medical Oncology, Università degli Studi di Verona, Verona, Italy. Running Title: Genomic Profiling in FGFR2-Rearranged Cholangiocarcinoma Keywords: pemigatinib, FGFR2, precision medicine, genomic profiling, cholangiocarcinoma Additional Information Financial Support: This study was sponsored by Incyte Corporation (Wilmington, Delaware). Corresponding Author: Timothy C. Burn, 1801 Augustine Cut-Off, Wilmington, DE 19803 W: 302 498 6787; [email protected] Conflict of Interest: 1 Downloaded from cancerdiscovery.aacrjournals.org on September 30, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on November 20, 2020; DOI: 10.1158/2159-8290.CD-20-0766 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. I.M. Silverman has employment with and owns stocks from Incyte Corporation. A. Hollebecque is a paid consultant for Debiopharm and Incyte Corporation. L. Friboulet received research funding from Debiopharm. S. Owens has employment with and owns stocks from Incyte Corporation. R.C.
    [Show full text]
  • CLIP2 As Radiation Biomarker in Papillary Thyroid Carcinoma
    Oncogene (2015) 34, 3917–3925 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc ORIGINAL ARTICLE CLIP2 as radiation biomarker in papillary thyroid carcinoma M Selmansberger1, A Feuchtinger2, L Zurnadzhy3, A Michna1, JC Kaiser4, M Abend5, A Brenner6, T Bogdanova3, A Walch2, K Unger1,7, H Zitzelsberger1,7 and J Hess1,7 A substantial increase in papillary thyroid carcinoma (PTC) among children exposed to the radioiodine fallout has been one of the main consequences of the Chernobyl reactor accident. Recently, the investigation of PTCs from a cohort of young patients exposed to the post-Chernobyl radioiodine fallout at very young age and a matched nonexposed control group revealed a radiation-specific DNA copy number gain on chromosomal band 7q11.23 and the radiation-associated mRNA overexpression of CLIP2. In this study, we investigated the potential role of CLIP2 as a radiation marker to be used for the individual classification of PTCs into CLIP2- positive and -negative cases—a prerequisite for the integration of CLIP2 into epidemiological modelling of the risk of radiation- induced PTC. We were able to validate the radiation-associated CLIP2 overexpression at the protein level by immunohistochemistry (IHC) followed by relative quantification using digital image analysis software (P = 0.0149). Furthermore, we developed a standardized workflow for the determination of CLIP2-positive and -negative cases that combines visual CLIP2 IHC scoring and CLIP2 genomic copy number status. In addition to the discovery cohort (n = 33), two independent validation cohorts of PTCs (n = 115) were investigated. High sensitivity and specificity rates for all three investigated cohorts were obtained, demonstrating robustness of the developed workflow.
    [Show full text]
  • Nguyen Thanh Binh National University of Singapore 2017
    COMPUTATIONAL STUDIES ON THE STRUCTURAL ASPECTS OF PROTEIN-PEPTIDE INTERACTIONS NGUYEN THANH BINH NATIONAL UNIVERSITY OF SINGAPORE 2017 COMPUTATIONAL STUDIES ON NGUYEN THANH BINH 2017 THE STRUCTURAL ASPECTS OF PROTEIN-PEPTIDE INTERACTIONS COMPUTATIONAL STUDIES ON THE STRUCTURAL ASPECTS OF PROTEIN-PEPTIDE INTERACTIONS NGUYEN THANH BINH (Master of Science, FSU, Russia) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF BIOLOGICAL SCIENCES NATIONAL UNIVERSITY OF SINGAPORE 2017 Supervisors: Associate Professor M. S. Madhusudhan, Main supervisor Dr Chandra Shekhar Verma, Co-supervisor Examiners: Associate Professor Kunchithapadam Swaminathan Assistant Professor Sebastian Maurer-Stroh Professor Ramasubbu Sankararamakrishnan, Indian Institute of Technology, Kanpur Declaration I hereby declare that this thesis is my original work and it has been written by me in its entirety. I have duly acknowledged all the sources of information that have been used in the thesis. This thesis has also not been submitted for any degree in any university previously. Nguyen Thanh Binh 12 Jan 2017 i Acknowledgments First I would like to thank my supervisor, Dr. M.S. Madhusudhan for all these years of mentoring and support. He has taught me not only bioinformatics, but also other scientific research skills, oral presentation and scientific writing. He has also given me valuable advices to overcome the stressful time of PhD program. I would like to thank my co-supervisor, Dr. Chandra Verma for his suggestion and guidance. I would like to thank my collaborator, Dr. Kim Chu Young for his insights into the research topics. Secondly, I would like to thank our group members, especially, Minh Nguyen, Kuan Pern Tan, Neelesh Soni, Shipra Gupta, Parichit Sharma and many others for their helps and discussions on various projects.
    [Show full text]
  • Expression Profiling of Human Milk Derived Exosomal Micrornas And
    www.nature.com/scientificreports OPEN Expression profling of human milk derived exosomal microRNAs and their targets in HIV‑1 infected mothers Muhammad Atif Zahoor1,2,8*, Xiao‑Dan Yao1,2, Bethany M. Henrick3,4, Chris P. Verschoor1,2,5, Alash’le Abimiku6,7, Sophia Osawe6 & Kenneth L. Rosenthal1,2* Despite the use of antiretroviral therapy (ART) in HIV‑1 infected mothers approximately 5% of new HIV‑1 infections still occur in breastfed infants annually, which warrants for the development of novel strategies to prevent new HIV‑1 infections in infants. Human milk (HM) exosomes are highly enriched in microRNAs (miRNAs), which play an important role in neonatal immunity. Furthermore, HM exosomes from healthy donors are known to inhibit HIV‑1 infection and transmission; however, the efect of HIV‑1 on HM exosomal miRNA signatures remains unknown. In this study, we used nCounter NanoString technology and investigated miRNAs expression profles in frst week postpartum HM exosomes from HIV‑1 infected and uninfected control mothers (n = 36). Our results indicated that HIV‑1 perturbed the diferential expression patterns of 19 miRNAs (13 upregulated and 6 downregulated) in HIV‑1 infected women compared to healthy controls. DIANA‑miR functional pathway analyses revealed that multiple biological pathways are involved including cell cycle, pathways in cancer, TGF‑β signaling, FoxO signaling, fatty acid biosynthesis, p53 signaling and apoptosis. Moreover, the receiver operating characteristics (ROC) curve analyses of miR‑630 and miR‑ 378g yielded areas under the ROC curves of 0.82 (95% CI 0.67 to 0.82) and 0.83 (95% CI 0.67 to 0.83), respectively highlighting their potential to serve as biomarkers to identify HIV‑1 infection in women.
    [Show full text]
  • Predicting Human Genes Susceptible to Genomic Instability Associated with Alu/Alu-Mediated Rearrangements
    Downloaded from genome.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press Predicting human genes susceptible to genomic instability associated with Alu/Alu-mediated rearrangements Key words: genome instability, Alu, genomic rearrangement, MMBIR, bioinformatics, machine- learning Xiaofei Song1, Christine R. Beck1, Renqian Du1, Ian M. Campbell1, Zeynep Coban-Akdemir1, Shen Gu1, Amy M. Breman1,2, Pawel Stankiewicz1,2, Grzegorz Ira1, Chad A. Shaw1,2, James R. Lupski1,3,4,5* 1. Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA 2. Baylor Genetics, Houston, TX 77021, USA 3. Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA 4. Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA 5. Texas Children’s Hospital, Houston, TX 77030, USA *Corresponding author: James R. Lupski, M.D., Ph.D., D.Sc. (hon), FAAP, FACMG, FANA, FAAAS, FAAS Cullen Professor, Department of Molecular and Human Genetics and Professor of Pediatrics Member, Human Genome Sequencing Center (HGSC) Baylor College of Medicine One Baylor Plaza Houston, Texas 77030 E-mail: [email protected] Phone: (713) 798-6530 Fax: (713) 798-5073 Downloaded from genome.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press Abstract Alu elements, the short interspersed element numbering >1 million copies per human genome, can mediate the formation of copy number variants (CNVs) between substrate pairs. These Alu/Alu-mediated rearrangements (AAMR) can result in pathogenic variants that cause diseases. To investigate the impact of AAMR on gene variation and human health, we first characterized Alus that are involved in mediating CNVs (CNV-Alus) and observed that these Alus tend to be evolutionarily younger.
    [Show full text]