Downloaded from the Gene Expression Omnibus (GSE25219) Were Used for the Assessment of Prenatal Gene Expression

Total Page:16

File Type:pdf, Size:1020Kb

Downloaded from the Gene Expression Omnibus (GSE25219) Were Used for the Assessment of Prenatal Gene Expression UCLA UCLA Electronic Theses and Dissertations Title Investigation of sex-differential genetic risk factors for autism spectrum disorders Permalink https://escholarship.org/uc/item/4cf1h61b Author Werling, Donna Marie Publication Date 2014 Peer reviewed|Thesis/dissertation eScholarship.org Powered by the California Digital Library University of California UNIVERSITY OF CALIFORNIA Los Angeles Investigation of sex-differential genetic risk factors for autism spectrum disorders A dissertation submitted in partial satisfaction of the requirements for the degree of Doctor of Philosophy in Neuroscience by Donna Marie Werling 2014 © Copyright by Donna Marie Werling 2014 ABSTRACT OF THE DISSERTATION Investigation of sex-differential genetic risk factors for autism spectrum disorders by Donna Marie Werling Doctor of Philosophy in Neuroscience University of California, Los Angeles, 2014 Professor Daniel H. Geschwind, Chair Autism spectrum disorders (ASDs) are pervasive neurodevelopmental disorders that affect more males than females, and the mechanisms responsible for increasing males’ risk or protecting females are not understood. This sex biased prevalence is consistent across time and populations, suggesting that an understanding of the processes driving sex-differential risk would likely be informative of fundamental pathophysiology in ASD. One known component of ASD risk is genetic variation. Thus, here I apply several approaches that leverage current knowledge of ASD genetics to investigate the role and mechanisms of sex-differential biology in ASD risk. First, I evaluate a cohort of families with more than one autistic child for evidence of sex- differential, familial risk variation. Second, I use genetic linkage analysis to identify sex- differential risk loci in families from the same multiplex cohort. Third, I characterize gene expression patterns in typical human neocortex to identify points of interaction between typical sexual dimorphism and genes known to carry risk variants for ASD. ii I find that recurrence rates for ASD diagnoses in multiplex families are consistent with a female protective model, in which females require more deleterious genetic variation to be affected with ASD and this greater genetic load is shared with females’ siblings. I also identify several chromosomal loci with evidence of genetic linkage in families either with (chromosome 8p21.2 and 8p12), or without (chromosome 1p31.3), an autistic female. No significant common variants are found in either region that can account for this linkage; these loci will be further investigated by targeted sequencing to identify rare risk variants. Gene expression analyses show that known ASD risk genes are not differentially expressed in males or females in the prenatal or adult human neocortex. However, astrocyte markers and gene sets implicated in immune function and inflammatory processes are expressed at higher levels in males. This suggests that sex-differential factors may operate downstream from, or interact with, ASD risk genes, as opposed to directly regulating the expression of these genes. Overall, findings from these multiple approaches provide valuable context for the function of sex-differential biology in ASD etiology, and suggest promising directions for future research. iii The dissertation of Donna Marie Werling is approved. Arthur P. Arnold Rita M. Cantor Nelson B. Freimer Daniel H. Geschwind, Committee Chair University of California, Los Angeles 2014 iv TABLE OF CONTENTS Chapter 1: Sex differences in autism spectrum disorders......................................................... 1 1.1: Introduction.................................................................................................................. 2 1.2: ASD prevalence in males and females........................................................................ 3 1.3: Presentation of ASD symptoms and related phenotypes in males and females.......... 5 1.4: Sex differences in genetic contributions to ASD risk.................................................. 7 1.5: Sex hormonal contributors to ASD risk....................................................................... 12 1.6: Conclusions.................................................................................................................. 16 Chapter 2: Recurrence rates provide evidence for sex-differential, familial genetic liability for autism spectrum disorders in multiplex families and twins...................................................... 18 2.1: Abstract........................................................................................................................ 19 2.2: Background.................................................................................................................. 20 2.3: Materials and methods................................................................................................. 24 2.3.a: Subjects............................................................................................................... 24 2.3.b: Sex ratios and recurrence risk............................................................................. 26 2.3.c: Quantitative phenotypes..................................................................................... 28 2.3.d: Concordance in twin pairs.................................................................................. 29 2.4: Results.......................................................................................................................... 30 2.5: Discussion.................................................................................................................... 39 2.6: Conclusions.................................................................................................................. 47 Chapter 3: Identification of suggestive sex-differential risk loci and replication of linkage at chromosome 20p13 for autism spectrum disorder.................................................................... 49 3.1: Abstract........................................................................................................................ 50 3.2: Background.................................................................................................................. 51 3.3: Materials and methods................................................................................................. 55 3.3.a: Subjects and genotyping..................................................................................... 55 3.3.b: Linkage analyses using all families.................................................................... 58 3.3.c: Sex-stratified linkage.......................................................................................... 59 3.3.d: Imputation........................................................................................................... 61 v 3.3.e: Linkage-directed association testing................................................................... 62 3.4: Results.......................................................................................................................... 63 3.4.a: Linkage in all families........................................................................................ 63 3.4.b: Sex-stratified linkage.......................................................................................... 66 3.4.c: Linkage-directed association.............................................................................. 72 3.5: Discussion.................................................................................................................... 73 3.6: Conclusions.................................................................................................................. 78 Chapter 4: Strategies for the identification of functional autism risk variants in linkage regions . .............................................................................................................................. 79 4.1: Linkage signals in autism spectrum disorders............................................................. 80 4.2: Common variant association........................................................................................ 82 4.3: Rare variant association............................................................................................... 84 4.4: Potential implications.................................................................................................. 89 Chapter 5: Gene expression implicates pathways at the interface between sexual dimorphisms and genetic risk variants for autism spectrum disorders........................................................... 90 5.1: Abstract........................................................................................................................ 91 5.2: Background.................................................................................................................. 91 5.3: Materials and methods................................................................................................. 96 5.3.a: Gene expression data from human brain tissue.................................................. 96 5.3.a.i: Adult BrainSpan sample............................................................................ 96 5.3.a.ii: Adult replication sample............................................................................ 97 5.3.a.iii: Prenatal BrainSpan sample....................................................................... 99 5.3.b: Differential
Recommended publications
  • Screening and Identification of Key Biomarkers in Clear Cell Renal Cell Carcinoma Based on Bioinformatics Analysis
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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. Screening and identification of key biomarkers in clear cell renal cell carcinoma based on bioinformatics analysis Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423889; this version posted December 23, 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 Clear cell renal cell carcinoma (ccRCC) is one of the most common types of malignancy of the urinary system. The pathogenesis and effective diagnosis of ccRCC have become popular topics for research in the previous decade. In the current study, an integrated bioinformatics analysis was performed to identify core genes associated in ccRCC. An expression dataset (GSE105261) was downloaded from the Gene Expression Omnibus database, and included 26 ccRCC and 9 normal kideny samples. Assessment of the microarray dataset led to the recognition of differentially expressed genes (DEGs), which was subsequently used for pathway and gene ontology (GO) enrichment analysis.
    [Show full text]
  • AP2A2 Antibody Cat
    AP2A2 Antibody Cat. No.: 63-513 AP2A2 Antibody Formalin-fixed and paraffin-embedded human Flow cytometric analysis of HepG2 cells using AP2A2 hepatocarcinoma with AP2A2 Antibody , which was Antibody (bottom histogram) compared to a negative peroxidase-conjugated to the secondary antibody, control cell (top histogram). FITC-conjugated goat-anti- followed by DAB staining. rabbit secondary antibodies were used for the analysis. Specifications HOST SPECIES: Rabbit SPECIES REACTIVITY: Human This AP2A2 antibody is generated from rabbits immunized with a KLH conjugated IMMUNOGEN: synthetic peptide between 610-637 amino acids from the Central region of human AP2A2. TESTED APPLICATIONS: Flow, IHC-P, WB For WB starting dilution is: 1:1000 APPLICATIONS: For IHC-P starting dilution is: 1:10~50 For FACS starting dilution is: 1:10~50 September 25, 2021 1 https://www.prosci-inc.com/ap2a2-antibody-63-513.html PREDICTED MOLECULAR 104 kDa WEIGHT: Properties This antibody is purified through a protein A column, followed by peptide affinity PURIFICATION: purification. CLONALITY: Polyclonal ISOTYPE: Rabbit Ig CONJUGATE: Unconjugated PHYSICAL STATE: Liquid BUFFER: Supplied in PBS with 0.09% (W/V) sodium azide. CONCENTRATION: batch dependent Store at 4˚C for three months and -20˚C, stable for up to one year. As with all antibodies STORAGE CONDITIONS: care should be taken to avoid repeated freeze thaw cycles. Antibodies should not be exposed to prolonged high temperatures. Additional Info OFFICIAL SYMBOL: AP2A2 AP-2 complex subunit alpha-2, 100 kDa coated vesicle
    [Show full text]
  • The Proximal Signaling Network of the BCR-ABL1 Oncogene Shows a Modular Organization
    Oncogene (2010) 29, 5895–5910 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 www.nature.com/onc ORIGINAL ARTICLE The proximal signaling network of the BCR-ABL1 oncogene shows a modular organization B Titz, T Low, E Komisopoulou, SS Chen, L Rubbi and TG Graeber Crump Institute for Molecular Imaging, Institute for Molecular Medicine, Jonsson Comprehensive Cancer Center, California NanoSystems Institute, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA BCR-ABL1 is a fusion tyrosine kinase, which causes signaling effects of BCR-ABL1 toward leukemic multiple types of leukemia. We used an integrated transformation. proteomic approach that includes label-free quantitative Oncogene (2010) 29, 5895–5910; doi:10.1038/onc.2010.331; protein complex and phosphorylation profiling by mass published online 9 August 2010 spectrometry to systematically characterize the proximal signaling network of this oncogenic kinase. The proximal Keywords: adaptor protein; BCR-ABL1; phospho- BCR-ABL1 signaling network shows a modular and complex; quantitative mass spectrometry; signaling layered organization with an inner core of three leukemia network; systems biology transformation-relevant adaptor protein complexes (Grb2/Gab2/Shc1 complex, CrkI complex and Dok1/ Dok2 complex). We introduced an ‘interaction direction- ality’ analysis, which annotates static protein networks Introduction with information on the directionality of phosphorylation- dependent interactions. In this analysis, the observed BCR-ABL1 is a constitutively active oncogenic fusion network structure was consistent with a step-wise kinase that arises through a chromosomal translocation phosphorylation-dependent assembly of the Grb2/Gab2/ and causes multiple types of leukemia. It is found in Shc1 and the Dok1/Dok2 complexes on the BCR-ABL1 many cases (B25%) of adult acute lymphoblastic core.
    [Show full text]
  • Genomic Modelling of Bipolar Disorders: Comparison of Multifactor Dimension Reduction and Classification-Based Data Mining Methods
    GENOMIC MODELLING OF BIPOLAR DISORDERS: COMPARISON OF MULTIFACTOR DIMENSION REDUCTION AND CLASSIFICATION-BASED DATA MINING METHODS A THESIS SUBMITTED TO THE GRADUATE SCHOOL OF INFORMATICS OF THE MIDDLE EAST TECHNICAL UNIVERSITY BY CENGİZHAN AÇIKEL IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY IN THE DEPARTMENT OF MEDICAL INFORMATICS MARCH 2017 GENOMIC MODELLING OF BIPOLAR DISORDERS: COMPARISON OF MULTIFACTOR DIMENSION REDUCTION AND CLASSIFICATION-BASED DATA MINING METHODS Submitted by CENGİZHAN AÇIKEL in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Department of Medical Informatics, Middle East Technical University by, Prof. Dr. Deniz Zeyrek Bozşahin ______________ Director, Graduate School of Informatics Assoc. Prof. Dr. Yeşim Aydın Son ______________ Head of Department, Health Informatics Assoc. Prof. Dr. Yeşim Aydın Son ______________ Supervisor, Health Informatics, METU Examining Committee Members: Assoc.Prof.Dr. Cem İyigün ______________ Industrial Engineering, METU Assoc. Prof. Dr. Yeşim Aydin Son ______________ Supervisor, Health Informatics, METU Assist. Prof. Dr. Aybar Can Acar ______________ Health Informatics, METU Assoc.Prof.Dr. Güvem Gümüş Akay ______________ Brain Research Center, Ankara University Assist.Prof.Dr. Ercüment Çiçek ______________ Computer Engineering, Bilkent University Date: ____/____/______ iii I hereby declare that all information in this document has been obtained and presented in accordance with academic rules and ethical conduct. I also declare that, as required by these rules and conduct, I have fully cited and referenced all material and results that are not original to this work. Name, Last Name : Cengizhan AÇIKEL Signature : __________________ iii ABSTRACT GENOMIC MODELLING OF BIPOLAR DISORDERS: COMPARISON OF MULTIFACTOR DIMENSION REDUCTION AND CLASSIFICATION-BASED DATA MINING METHODS Açıkel, Cengizhan Ph.D.
    [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]
  • Multivariate Meta-Analysis of Differential Principal Components Underlying Human Primed and Naive-Like Pluripotent States
    bioRxiv preprint doi: https://doi.org/10.1101/2020.10.20.347666; this version posted October 21, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. October 20, 2020 To: bioRxiv Multivariate Meta-Analysis of Differential Principal Components underlying Human Primed and Naive-like Pluripotent States Kory R. Johnson1*, Barbara S. Mallon2, Yang C. Fann1, and Kevin G. Chen2*, 1Intramural IT and Bioinformatics Program, 2NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA Keywords: human pluripotent stem cells; naive pluripotency, meta-analysis, principal component analysis, t-SNE, consensus clustering *Correspondence to: Dr. Kory R. Johnson ([email protected]) Dr. Kevin G. Chen ([email protected]) 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.10.20.347666; this version posted October 21, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. ABSTRACT The ground or naive pluripotent state of human pluripotent stem cells (hPSCs), which was initially established in mouse embryonic stem cells (mESCs), is an emerging and tentative concept. To verify this important concept in hPSCs, we performed a multivariate meta-analysis of major hPSC datasets via the combined analytic powers of percentile normalization, principal component analysis (PCA), t-distributed stochastic neighbor embedding (t-SNE), and SC3 consensus clustering.
    [Show full text]
  • Host-Pathogen Systems Biology: Logical Modelling of Hepatocyte
    BMC Systems Biology BioMed Central Research article Open Access Host-pathogen systems biology: logical modelling of hepatocyte growth factor and Helicobacter pylori induced c-Met signal transduction Raimo Franke†1, Melanie Müller†1, Nicole Wundrack1, Ernst-Dieter Gilles2, Steffen Klamt2, Thilo Kähne1 and Michael Naumann*1 Address: 1Institute of Experimental Internal Medicine, Otto von Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany and 2Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany Email: Raimo Franke - [email protected]; Melanie Müller - [email protected]; Nicole Wundrack - [email protected]; Ernst-Dieter Gilles - [email protected]; Steffen Klamt - Klamt@mpi- magdeburg.mpg.de; Thilo Kähne - [email protected]; Michael Naumann* - [email protected] * Corresponding author †Equal contributors Published: 14 January 2008 Received: 3 October 2007 Accepted: 14 January 2008 BMC Systems Biology 2008, 2:4 doi:10.1186/1752-0509-2-4 This article is available from: http://www.biomedcentral.com/1752-0509/2/4 © 2008 Franke et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Background: The hepatocyte growth factor (HGF) stimulates mitogenesis, motogenesis, and morphogenesis in a wide range of tissues, including epithelial cells, on binding to the receptor tyrosine kinase c-Met. Abnormal c-Met signalling contributes to tumour genesis, in particular to the development of invasive and metastatic phenotypes.
    [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]
  • RET Gene Fusions in Malignancies of the Thyroid and Other Tissues
    G C A T T A C G G C A T genes Review RET Gene Fusions in Malignancies of the Thyroid and Other Tissues Massimo Santoro 1,*, Marialuisa Moccia 1, Giorgia Federico 1 and Francesca Carlomagno 1,2 1 Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Naples, Italy; [email protected] (M.M.); [email protected] (G.F.); [email protected] (F.C.) 2 Institute of Endocrinology and Experimental Oncology of the CNR, 80131 Naples, Italy * Correspondence: [email protected] Received: 10 March 2020; Accepted: 12 April 2020; Published: 15 April 2020 Abstract: Following the identification of the BCR-ABL1 (Breakpoint Cluster Region-ABelson murine Leukemia) fusion in chronic myelogenous leukemia, gene fusions generating chimeric oncoproteins have been recognized as common genomic structural variations in human malignancies. This is, in particular, a frequent mechanism in the oncogenic conversion of protein kinases. Gene fusion was the first mechanism identified for the oncogenic activation of the receptor tyrosine kinase RET (REarranged during Transfection), initially discovered in papillary thyroid carcinoma (PTC). More recently, the advent of highly sensitive massive parallel (next generation sequencing, NGS) sequencing of tumor DNA or cell-free (cfDNA) circulating tumor DNA, allowed for the detection of RET fusions in many other solid and hematopoietic malignancies. This review summarizes the role of RET fusions in the pathogenesis of human cancer. Keywords: kinase; tyrosine kinase inhibitor; targeted therapy; thyroid cancer 1. The RET Receptor RET (REarranged during Transfection) was initially isolated as a rearranged oncoprotein upon the transfection of a human lymphoma DNA [1].
    [Show full text]
  • Identification of Potential Key Genes and Pathway Linked with Sporadic Creutzfeldt-Jakob Disease Based on Integrated Bioinformatics Analyses
    medRxiv preprint doi: https://doi.org/10.1101/2020.12.21.20248688; this version posted December 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Identification of potential key genes and pathway linked with sporadic Creutzfeldt-Jakob disease based on integrated bioinformatics analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 , Iranna Kotturshetti 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. 3. Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka 562209, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice. medRxiv preprint doi: https://doi.org/10.1101/2020.12.21.20248688; this version posted December 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Abstract Sporadic Creutzfeldt-Jakob disease (sCJD) is neurodegenerative disease also called prion disease linked with poor prognosis. The aim of the current study was to illuminate the underlying molecular mechanisms of sCJD. The mRNA microarray dataset GSE124571 was downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were screened.
    [Show full text]
  • Imatinib Dependent Tyrosine Phosphorylation Profiling of Bcr-Abl
    Provided by the author(s) and University College Dublin Library in accordance with publisher policies. Please cite the published version when available. Title Imatinib-dependent tyrosine phosphorylation profiling of Bcr-Abl-positive chronic myeloid leukemia cells Authors(s) Preisinger, C.; Schwarz, J. P.; Bleijerveld, O. B.; et al. Publication date 2012-08-27 Publication information Leukemia, 27 (3): 743-746 Publisher Nature Publishing Group Item record/more information http://hdl.handle.net/10197/5078 Publisher's version (DOI) 10.1038/leu.2012.243 Downloaded 2021-09-25T14:43:28Z The UCD community has made this article openly available. Please share how this access benefits you. Your story matters! (@ucd_oa) © Some rights reserved. For more information, please see the item record link above. 1 Imatinib dependent tyrosine phosphorylation profiling of Bcr-Abl 2 positive chronic myeloid leukemia cells 3 Bcr-Abl is the major cause and pathogenetic principle of chronic myeloid leukemia (CML). 4 Bcr-Abl results from a chromosomal translocation that fuses the bcr and the abl genes, 5 thereby generating a constitutively active tyrosine kinase, which stimulates several signaling 6 networks required for proliferation and survival. Bcr-Abl’s oncogenic properties comprise 7 both functions as a kinase and as a scaffold protein 1. A number of Bcr-Abl interaction 8 partners and downstream effectors have been described, improving our understanding of the 9 signaling networks deranged in CML. Brehme et al. recently defined the “core-interactome of 10 Bcr-Abl”, identifying seven major interaction partners of Bcr-Abl (GRB2, Shc1, Crk, c-Cbl, 11 p85, Sts-1, and SHIP-2) 2.
    [Show full text]
  • Literature Mining Sustains and Enhances Knowledge Discovery from Omic Studies
    LITERATURE MINING SUSTAINS AND ENHANCES KNOWLEDGE DISCOVERY FROM OMIC STUDIES by Rick Matthew Jordan B.S. Biology, University of Pittsburgh, 1996 M.S. Molecular Biology/Biotechnology, East Carolina University, 2001 M.S. Biomedical Informatics, University of Pittsburgh, 2005 Submitted to the Graduate Faculty of School of Medicine in partial fulfillment of the requirements for the degree of Doctor of Philosophy University of Pittsburgh 2016 UNIVERSITY OF PITTSBURGH SCHOOL OF MEDICINE This dissertation was presented by Rick Matthew Jordan It was defended on December 2, 2015 and approved by Shyam Visweswaran, M.D., Ph.D., Associate Professor Rebecca Jacobson, M.D., M.S., Professor Songjian Lu, Ph.D., Assistant Professor Dissertation Advisor: Vanathi Gopalakrishnan, Ph.D., Associate Professor ii Copyright © by Rick Matthew Jordan 2016 iii LITERATURE MINING SUSTAINS AND ENHANCES KNOWLEDGE DISCOVERY FROM OMIC STUDIES Rick Matthew Jordan, M.S. University of Pittsburgh, 2016 Genomic, proteomic and other experimentally generated data from studies of biological systems aiming to discover disease biomarkers are currently analyzed without sufficient supporting evidence from the literature due to complexities associated with automated processing. Extracting prior knowledge about markers associated with biological sample types and disease states from the literature is tedious, and little research has been performed to understand how to use this knowledge to inform the generation of classification models from ‘omic’ data. Using pathway analysis methods to better understand the underlying biology of complex diseases such as breast and lung cancers is state-of-the-art. However, the problem of how to combine literature- mining evidence with pathway analysis evidence is an open problem in biomedical informatics research.
    [Show full text]