The Role of Bip Co-Chaperone SIL1 in Marinesco-Sjögren Syndrome Pathogenesis" (2018)

Total Page:16

File Type:pdf, Size:1020Kb

The Role of Bip Co-Chaperone SIL1 in Marinesco-Sjögren Syndrome Pathogenesis University of Tennessee Health Science Center UTHSC Digital Commons Theses and Dissertations (ETD) College of Graduate Health Sciences 12-2018 The Role of BiP Co-chaperone SIL1 in Marinesco- Sjögren Syndrome Pathogenesis Viraj Paresh Ichhaporia University of Tennessee Health Science Center Follow this and additional works at: https://dc.uthsc.edu/dissertations Part of the Medical Cell Biology Commons, and the Medical Molecular Biology Commons Recommended Citation Ichhaporia, Viraj Paresh (http://orcid.org/0000-0001-7466-6487), "The Role of BiP Co-chaperone SIL1 in Marinesco-Sjögren Syndrome Pathogenesis" (2018). Theses and Dissertations (ETD). Paper 469. http://dx.doi.org/10.21007/etd.cghs.2018.0470. This Dissertation is brought to you for free and open access by the College of Graduate Health Sciences at UTHSC Digital Commons. It has been accepted for inclusion in Theses and Dissertations (ETD) by an authorized administrator of UTHSC Digital Commons. For more information, please contact [email protected]. The Role of BiP Co-chaperone SIL1 in Marinesco-Sjögren Syndrome Pathogenesis Document Type Dissertation Degree Name Doctor of Philosophy (PhD) Program Biomedical Sciences Track Microbiology, Immunology, and Biochemistry Research Advisor Linda M. Hendershot, Ph.D. Committee Alessandra d'Azzo, Ph.D. Tony N. Marion, Ph.D. David R. Nelson, Ph.D. J. Paul Taylor, M.D., Ph.D. Stanislav S. Zakharenko, M.D., Ph.D. ORCID http://orcid.org/0000-0001-7466-6487 DOI 10.21007/etd.cghs.2018.0470 This dissertation is available at UTHSC Digital Commons: https://dc.uthsc.edu/dissertations/469 The Role of BiP Co-chaperone SIL1 in Marinesco-Sjögren Syndrome Pathogenesis A Dissertation Presented for The Graduate Studies Council The University of Tennessee Health Science Center In Partial Fulfillment Of the Requirements for the Degree Doctor of Philosophy From The University of Tennessee By Viraj Paresh Ichhaporia December 2018 Chapter 2 © 2015 by Viraj P. Ichhaporia et al. All other material © 2018 by Viraj P. Ichhaporia. All rights reserved. ii DEDICATION Dedicated to my parents and my grandparents. It is because of their countless sacrifices, unconditional love, and everlasting support that have inspired me to push beyond my personal limits, and who have instilled in me the values that I most cherish today. iii ACKNOWLEDGEMENTS It has been an incredible journey over the past seven years, studded with numerous memories and learning lessons, that brings me to the end of my dissertation. Right from my lab rotation to until now, I have never stopped learning from Dr. Linda M. Hendershot, my Ph.D. advisor, and I am indebted to her for that. Dr. Hendershot has been an excellent mentor over these years and is responsible for inculcating in me the core values of an ideal scientist, while always being supportive and enabling me to be a better version of myself, at a professional and a personal level. I am grateful to her for giving me the opportunity to be a part of the family at Hendershot laboratory and the cherished memories that I carry forward. I shall always remember what I have learnt here. Along with Dr. Hendershot, I am very thankful to Melissa Mann for teaching me the rigor and prowess with which scientific experimentation should be performed. It is Ms. Mann who taught me many of the lab techniques that I proudly claim as my ‘bread and butter’ skills. I also wish to thank her for her help with numerous aspects of my dissertation research. I am also thankful to my friends within the Hendershot lab, both past and present, for enlivening the work place and for always being supportive, encouraging, and helpful at any given opportunity. It is only because of them that the late nights and long hours passed by with ease. I thank Tyler Sanford for early discussions on the potential of this project and helping me transition into this research field. I thank Drs. Brian Murphy and Walid Awad for their assistance with managing the mouse colony and genotyping. I truly appreciate the support and constructive feedback I have received over the years from Dr. Kyung Tae Chung (who discovered mammalian SIL1), Dr. Joel Otero, Dr. Ethel Pereira, Dr. Matthias Feige, Dr. Julia Behnke, Dr. Jyoti Sinha, Dr. Greg Poet, Dr. Kristine Pobre, Dr. Amanda Preston, and Mary Carson Irvine. A special thanks to my friends and fellow graduate students, Christina Oikonomou and Rachael Wood, for their wholehearted support. I wish them the best for their own Ph.D. journeys. I was fortunate to work with an excellent summer student and a budding young scientist, Jessica Joyce, and thank her for her help with the SIL1 mutants project. I sincerely also thank my committee members, Dr. Alessandra d’Azzo, Dr. Tony Marion, Dr. David Nelson, Dr. J. Paul Taylor, and Dr. Stanislav Zakharenko, for their time, advice, guidance, and support over these years. The decision to pursue proteomics and many riveting answers stemmed from the scientific discussions during my committee meetings. I would like to thank Dr. Alessandra d’Azzo, Dr. Mondira Kundu, Dr. J. Paul Taylor, and Dr. Bo Wang for their generous gift of reagents, and Dr. Diantha Van de Vlekkert and Dr. Cath Drummond for their help with establishing myoblast cultures. Without the help from my collaborators, answering questions using technologies that lie beyond my expertise would not have been possible. In doing so, these collaborators not only offered me their time, but also excellent opportunities to educate myself and understand complex data in a meaningful manner. I thank Sharon Frase and Linda Horner for their assistance with electron microscopy studies; Dr. Peter Vogel, Sean iv Savage, Michael Anderson, Michael Straign, and Chandra Savage for their help animal pathology and pharmacokinetic studies; Dr. Junmin Peng, Dr. Vishwajeeth Pagala, Dr. Kanisha Kavdia, and Dr. Yong-Dong Wang for their help with proteomic studies; Dr. Jieun Kim and Dr. Walter Akers for their help with PET-μCT studies; Dr. Chunliang Li and Shaela Wright for making the CRISPR-mediated SIL1 knockout cell lines a possibility; Yumei Zhang for all her help with the structural analysis of the SIL1 mutants, and Dr. Fabio Demontis for providing critical feedback and improving our SIL1- myopathy article. I would like to acknowledge the funding support I have received from UTHSC and St. Jude in the form of travel grants that have enabled me to present my research on diverse platforms and engage in career development activities. For this, I thank Dr. Donald Thomason, Dr. Gerard Zambetti, and Dr. Isaac Donkor. I thank NIH and ALSAC for their funding support through Dr. Hendershot that has allowed me to perform my experimentation. I also thank the UTHSC deans along with the past and present members of the Graduate Student Executive Committee for empowering and supporting graduate students. I also appreciate the help I have received from the faculty and staff members at UTHSC and St. Jude. In particular, I would like to call out the current and past IBS Program Directors, Dr. Elizabeth Fitzpatrick, Dr. Rennolds Ostrom, and Dr. Pat Ryan; and Ms. Elizabeth Webb, Late Ms. Tricia Satkowski, and Ms. Denise Slaughter, our past and present program coordinators from UTHSC and St. Jude. I also thank the faculty members of the Microbiology, Biochemistry, and Immunology department, and my colleagues within the Tumor Cell Biology department for their scientific feedback and guidance. Lastly, I am grateful beyond measure to my family, especially my parents, without whom this life-changing journey would have never materialized, and for their ever-lasting support and inspiration. I am very thankful to my fiancé for being my northern star and my strength. Last, but certainly not the least, I would like to end by thanking my friends who made my time in Memphis a truly enjoyable and nostalgic experience. v ABSTRACT Marinesco-Sjögren syndrome (MSS) is a rare, autosomal recessive, multisystem disorder, which is characterized by cerebellar ataxia, early-onset bilateral cataracts, and progressive myopathy amongst other symptoms. MSS has been attributed to mutations in the SIL1 gene, which encodes a nucleotide exchange factor for the endoplasmic- reticulum-resident Hsp70 chaperone, BiP. To date, there are 46 MSS-associated mutations that have been reported in SIL1, which occur throughout this gene and are predicted to result in a loss of SIL1’s function. The large majority of these mutations cause deletions of large fractions of the SIL1 protein. Nine MSS-associated mutations are particularly interesting because they alter less than six amino acids, yet are associated with a phenotype indistinguishable from a near-full length deletion of SIL1. The mechanisms by which these nine mutations lead to a loss of SIL1’s function are not well understood. On the other hand, it remains unclear how the loss of SIL1 leads to the multisystem defects observed in MSS, selectively affecting certain tissues while sparing others. Our goal was to answer these two questions. We have shown that the selected nine MSS-associated SIL1 mutations may dramatically alter the protein microenvironment and disrupt intramolecular interactions, such that it alters the folding properties of SIL1 and renders it aggregation-prone. This offers a potential mechanism by which mutations in SIL1 cause a loss of its function. We validated that the C57BL/6 Sil1Gt mouse model, which harbors a genetic disruption of Sil1, phenocopies numerous aspects of the MSS-phenotype and represents a valid preclinical model system to investigate the MSS-associated pathology and explore pharmacotherapeutic strategies. Using a combination of the Sil1Gt mice and SIL1- deficient MSS-patient-derived lymphoblastoid cell lines, we explored the biosynthesis and secretion of immunoglobulins (Ig), which are the best characterized substrate of BiP to date. In vivo antigen-specific immunizations and ex vivo LPS stimulation of splenic B cells revealed that the Sil1Gt mouse was indistinguishable from wild-type age-matched controls, in terms of both the kinetics and magnitude of antigen-specific antibody responses.
Recommended publications
  • 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]
  • Cldn19 Clic2 Clmp Cln3
    NewbornDx™ Advanced Sequencing Evaluation When time to diagnosis matters, the NewbornDx™ Advanced Sequencing Evaluation from Athena Diagnostics delivers rapid, 5- to 7-day results on a targeted 1,722-genes. A2ML1 ALAD ATM CAV1 CLDN19 CTNS DOCK7 ETFB FOXC2 GLUL HOXC13 JAK3 AAAS ALAS2 ATP1A2 CBL CLIC2 CTRC DOCK8 ETFDH FOXE1 GLYCTK HOXD13 JUP AARS2 ALDH18A1 ATP1A3 CBS CLMP CTSA DOK7 ETHE1 FOXE3 GM2A HPD KANK1 AASS ALDH1A2 ATP2B3 CC2D2A CLN3 CTSD DOLK EVC FOXF1 GMPPA HPGD K ANSL1 ABAT ALDH3A2 ATP5A1 CCDC103 CLN5 CTSK DPAGT1 EVC2 FOXG1 GMPPB HPRT1 KAT6B ABCA12 ALDH4A1 ATP5E CCDC114 CLN6 CUBN DPM1 EXOC4 FOXH1 GNA11 HPSE2 KCNA2 ABCA3 ALDH5A1 ATP6AP2 CCDC151 CLN8 CUL4B DPM2 EXOSC3 FOXI1 GNAI3 HRAS KCNB1 ABCA4 ALDH7A1 ATP6V0A2 CCDC22 CLP1 CUL7 DPM3 EXPH5 FOXL2 GNAO1 HSD17B10 KCND2 ABCB11 ALDOA ATP6V1B1 CCDC39 CLPB CXCR4 DPP6 EYA1 FOXP1 GNAS HSD17B4 KCNE1 ABCB4 ALDOB ATP7A CCDC40 CLPP CYB5R3 DPYD EZH2 FOXP2 GNE HSD3B2 KCNE2 ABCB6 ALG1 ATP8A2 CCDC65 CNNM2 CYC1 DPYS F10 FOXP3 GNMT HSD3B7 KCNH2 ABCB7 ALG11 ATP8B1 CCDC78 CNTN1 CYP11B1 DRC1 F11 FOXRED1 GNPAT HSPD1 KCNH5 ABCC2 ALG12 ATPAF2 CCDC8 CNTNAP1 CYP11B2 DSC2 F13A1 FRAS1 GNPTAB HSPG2 KCNJ10 ABCC8 ALG13 ATR CCDC88C CNTNAP2 CYP17A1 DSG1 F13B FREM1 GNPTG HUWE1 KCNJ11 ABCC9 ALG14 ATRX CCND2 COA5 CYP1B1 DSP F2 FREM2 GNS HYDIN KCNJ13 ABCD3 ALG2 AUH CCNO COG1 CYP24A1 DST F5 FRMD7 GORAB HYLS1 KCNJ2 ABCD4 ALG3 B3GALNT2 CCS COG4 CYP26C1 DSTYK F7 FTCD GP1BA IBA57 KCNJ5 ABHD5 ALG6 B3GAT3 CCT5 COG5 CYP27A1 DTNA F8 FTO GP1BB ICK KCNJ8 ACAD8 ALG8 B3GLCT CD151 COG6 CYP27B1 DUOX2 F9 FUCA1 GP6 ICOS KCNK3 ACAD9 ALG9
    [Show full text]
  • 2021 Code Changes Reference Guide
    Boston University Medical Group 2021 CPT Code Changes Reference Guide Page 1 of 51 Background Current Procedural Terminology (CPT) was created by the American Medical Association (AMA) in 1966. It is designed to be a means of effective and dependable communication among physicians, patients, and third-party payers. CPT provides a uniform coding scheme that accurately describes medical, surgical, and diagnostic services. CPT is used for public and private reimbursement systems; development of guidelines for medical care review; as a basis for local, regional, and national utilization comparisons; and medical education and research. CPT Category I codes describe procedures and services that are consistent with contemporary medical practice. Category I codes are five-digit numeric codes. CPT Category II codes facilitate data collection for certain services and test results that contribute to positive health outcomes and quality patient care. These codes are optional and used for performance management. They are alphanumeric five-digit codes with the alpha character F in the last position. CPT Category III codes represent emerging technologies. They are alphanumeric five-digit codes with the alpha character T in the last position. The CPT Editorial Panel, appointed by the AMA Board of Trustees, is responsible for maintaining and updating the CPT code set. Purpose The AMA makes annual updates to the CPT code set, effective January 1. These updates include deleted codes, revised codes, and new codes. It’s important for providers to understand the code changes and the impact those changes will have to systems, workflow, reimbursement, and RVUs. This document is meant to assist you with this by providing a summary of the changes; a detailed breakdown of this year’s CPT changes by specialty, and HCPCS Updates for your reference.
    [Show full text]
  • 158273472.Pdf
    ANNUAL .2003REPCOLD SPRING HARBOR LABORATORY .1; ANNUAL REPORT 2003 © 2004 by Cold Spring Harbor Laboratory Cold Spring Harbor Laboratory One Bungtown Road Cold Spring Harbor, New York 11724 Web Site: www.cshl.edu Managing Editors Jeff Picarello, Lisa Becker Production Editor Rena Steuer Copy Editor Dorothy Brown Development Manager Jan Argentine Project Coordinators Maria Falasca, Nora Rice Production Manager Denise Weiss Desktop Editor Susan Schaefer Nonscientific Photography Miriam Chua, Bill Geddes Cover Designer Denise Weiss Book Designer Emily Harste Front cover: McClintock Laboratory (right) and Carnegie Library (left) (photos by Miriam Chua) Back cover: Magnolia Kobus on grounds of Cold Spring Harbor Laboratory (photo by Bruce Stillman) Section title pages: Miriam Chua Contents Officers of the Corporation/Board of Trusteesiv-v Governancevi Committees vii Edwin Marks (1926-2003) viii PRESIDENT'S REPORT Highlights5 CHIEF OPERATING OFFICER'S REPORT 25 50TH ANNIVERSARY OF THE DOUBLE HELIX 29 RESEARCH 47 Cancer: Gene Expression 49 Cancer: Genetics 74 Cancer: Cell Biology 106 Bioinformatics and Genomics 134 Neuroscience152 Plant Development and Genetics 199 CSHL Fellows 212 Author Index 217 WATSON SCHOOL OF BIOLOGICAL SCIENCES 219 Dean's Report 221 Courses 238 Undergraduate Research Program245 Partners for the Future 248 Nature Study 249 COLD SPRING HARBOR LABORATORY MEETINGS AND COURSES 251 Academic Affairs253 Symposium on Quantitative Biology 255 Meetings 258 Postgraduate Courses295 Seminars 353 BANBURY CENTER 355 Director's Report357 Meetings 365 DOLAN DNA LEARNING CENTER 403 Director's Report 405 Workshops, Meetings, and Collaborations 418 COLD SPRING HARBOR LABORATORY PRESS 425 Publications 426 Executive Director's Report 427 FINANCE 431 History of the CSHL Endowment 433 Financial Statements 444 Financial Support448 Grants448 Institutional Advancement 457 Capital and Program Contributions 458 Watson School of Biological Sciences Capital Campaign 459 Annual Contributions 460 LABORATORY STAFF 474 III Officers of the Corporation William R.
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • 1 Fibrillar Αβ Triggers Microglial Proteome Alterations and Dysfunction in Alzheimer Mouse 1 Models 2 3 4 Laura Sebastian
    bioRxiv preprint doi: https://doi.org/10.1101/861146; this version posted December 2, 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 Fibrillar triggers microglial proteome alterations and dysfunction in Alzheimer mouse 2 models 3 4 5 Laura Sebastian Monasor1,10*, Stephan A. Müller1*, Alessio Colombo1, Jasmin König1,2, Stefan 6 Roth3, Arthur Liesz3,4, Anna Berghofer5, Takashi Saito6,7, Takaomi C. Saido6, Jochen Herms1,4,8, 7 Michael Willem9, Christian Haass1,4,9, Stefan F. Lichtenthaler 1,4,5# & Sabina Tahirovic1# 8 9 1 German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany 10 2 Faculty of Chemistry, Technical University of Munich, Garching, Germany 11 3 Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians Universität München, 12 81377 Munich, Germany 13 4 Munich Cluster for Systems Neurology (SyNergy), Munich, Germany 14 5 Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, 15 Munich, Germany 16 6 Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science Institute, Wako, 17 Saitama 351-0198, Japan 18 7 Department of Neurocognitive Science, Nagoya City University Graduate School of Medical 19 Science, Nagoya, Aichi 467-8601, Japan 20 8 Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität München, 81377 21 Munich, Germany 22 9 Biomedical Center (BMC), Ludwig-Maximilians Universität München, 81377 Munich, Germany 23 10 Graduate School of Systemic Neuroscience, Ludwig-Maximilians-University Munich, Munich, 24 Germany. 25 *Contributed equally 26 #Correspondence: [email protected] and [email protected] 27 28 Running title: 29 Microglial proteomic signatures of AD 30 Keywords: Alzheimer’s disease / microglia / proteomic signatures / neuroinflammation / 31 phagocytosis 32 1 bioRxiv preprint doi: https://doi.org/10.1101/861146; this version posted December 2, 2019.
    [Show full text]
  • Protein Symbol Protein Name Rank Metric Score 4F2 4F2 Cell-Surface
    Supplementary Table 2 Supplementary Table 2. Ranked list of proteins present in anti-Sema4D treated macrophage conditioned media obtained in the GSEA analysis of the proteomic data. Proteins are listed according to their rank metric score, which is the score used to position the gene in the ranked list of genes of the GSEA. Values are obtained from comparing Sema4D treated RAW conditioned media versus REST, which includes untreated, IgG treated and anti-Sema4D added RAW conditioned media. GSEA analysis was performed under standard conditions in November 2015. Protein Rank metric symbol Protein name score 4F2 4F2 cell-surface antigen heavy chain 2.5000 PLOD3 Procollagen-lysine,2-oxoglutarate 5-dioxygenase 3 1.4815 ELOB Transcription elongation factor B polypeptide 2 1.4350 ARPC5 Actin-related protein 2/3 complex subunit 5 1.2603 OSTF1 teoclast-stimulating factor 1 1.2500 RL5 60S ribomal protein L5 1.2135 SYK Lysine--tRNA ligase 1.2135 RL10A 60S ribomal protein L10a 1.2135 TXNL1 Thioredoxin-like protein 1 1.1716 LIS1 Platelet-activating factor acetylhydrolase IB subunit alpha 1.1067 A4 Amyloid beta A4 protein 1.0911 H2B1M Histone H2B type 1-M 1.0514 UB2V2 Ubiquitin-conjugating enzyme E2 variant 2 1.0381 PDCD5 Programmed cell death protein 5 1.0373 UCHL3 Ubiquitin carboxyl-terminal hydrolase isozyme L3 1.0061 PLEC Plectin 1.0061 ITPA Inine triphphate pyrophphatase 0.9524 IF5A1 Eukaryotic translation initiation factor 5A-1 0.9314 ARP2 Actin-related protein 2 0.8618 HNRPL Heterogeneous nuclear ribonucleoprotein L 0.8576 DNJA3 DnaJ homolog subfamily
    [Show full text]
  • Figure S1. GO Analysis of Genes in Glioblastoma Cases That Showed Positive and Negative Correlations with TCIRG1 in the GSE16011 Cohort
    Figure S1. GO analysis of genes in glioblastoma cases that showed positive and negative correlations with TCIRG1 in the GSE16011 cohort. (A‑C) GO‑BP, GO‑CC and GO‑MF terms of genes that showed positive correlations with TCIRG1, respec‑ tively. (D‑F) GO‑BP, GO‑CC and GO‑MF terms of genes that showed negative correlations with TCIRG1. Red nodes represent gene counts, and black bars represent negative 1og10 P‑values. TCIRG1, T cell immune regulator 1; GO, Gene Ontology; BP, biological process; CC, cellular component; MF, molecular function. Table SI. Genes correlated with T cell immune regulator 1. Gene Name Pearson's r ARPC1B Actin‑related protein 2/3 complex subunit 1B 0.756 IL4R Interleukin 4 receptor 0.695 PLAUR Plasminogen activator, urokinase receptor 0.693 IFI30 IFI30, lysosomal thiol reductase 0.675 TNFAIP3 TNF α‑induced protein 3 0.675 RBM47 RNA binding motif protein 47 0.666 TYMP Thymidine phosphorylase 0.665 CEBPB CCAAT/enhancer binding protein β 0.663 MVP Major vault protein 0.660 BCL3 B‑cell CLL/lymphoma 3 0.657 LILRB3 Leukocyte immunoglobulin‑like receptor B3 0.656 ELF4 E74 like ETS transcription factor 4 0.652 ITGA5 Integrin subunit α 5 0.651 SLAMF8 SLAM family member 8 0.647 PTPN6 Protein tyrosine phosphatase, non‑receptor type 6 0.641 RAB27A RAB27A, member RAS oncogene family 0.64 S100A11 S100 calcium binding protein A11 0.639 CAST Calpastatin 0.638 EHBP1L1 EH domain‑binding protein 1‑like 1 0.638 LILRB2 Leukocyte immunoglobulin‑like receptor B2 0.629 ALDH3B1 Aldehyde dehydrogenase 3 family member B1 0.626 GNA15 G protein
    [Show full text]
  • Genetic Testing Medical Policy – Genetics
    Genetic Testing Medical Policy – Genetics Please complete all appropriate questions fully. Suggested medical record documentation: • Current History & Physical • Progress Notes • Family Genetic History • Genetic Counseling Evaluation *Failure to include suggested medical record documentation may result in delay or possible denial of request. PATIENT INFORMATION Name: Member ID: Group ID: PROCEDURE INFORMATION Genetic Counseling performed: c Yes c No **Please check the requested analyte(s), identify number of units requested, and provide indication/rationale for testing. 81400 Molecular Pathology Level 1 Units _____ c ACADM (acyl-CoA dehydrogenase, C-4 to C-12 straight chain, MCAD) (e.g., medium chain acyl dehydrogenase deficiency), K304E variant _____ c ACE (angiotensin converting enzyme) (e.g., hereditary blood pressure regulation), insertion/deletion variant _____ c AGTR1 (angiotensin II receptor, type 1) (e.g., essential hypertension), 1166A>C variant _____ c BCKDHA (branched chain keto acid dehydrogenase E1, alpha polypeptide) (e.g., maple syrup urine disease, type 1A), Y438N variant _____ c CCR5 (chemokine C-C motif receptor 5) (e.g., HIV resistance), 32-bp deletion mutation/794 825del32 deletion _____ c CLRN1 (clarin 1) (e.g., Usher syndrome, type 3), N48K variant _____ c DPYD (dihydropyrimidine dehydrogenase) (e.g., 5-fluorouracil/5-FU and capecitabine drug metabolism), IVS14+1G>A variant _____ c F13B (coagulation factor XIII, B polypeptide) (e.g., hereditary hypercoagulability), V34L variant _____ c F2 (coagulation factor 2) (e.g.,
    [Show full text]
  • Perkinelmer Genomics to Request the Saliva Swab Collection Kit for Patients That Cannot Provide a Blood Sample As Whole Blood Is the Preferred Sample
    Autism and Intellectual Disability TRIO Panel Test Code TR002 Test Summary This test analyzes 2429 genes that have been associated with Autism and Intellectual Disability and/or disorders associated with Autism and Intellectual Disability with the analysis being performed as a TRIO Turn-Around-Time (TAT)* 3 - 5 weeks Acceptable Sample Types Whole Blood (EDTA) (Preferred sample type) DNA, Isolated Dried Blood Spots Saliva Acceptable Billing Types Self (patient) Payment Institutional Billing Commercial Insurance Indications for Testing Comprehensive test for patients with intellectual disability or global developmental delays (Moeschler et al 2014 PMID: 25157020). Comprehensive test for individuals with multiple congenital anomalies (Miller et al. 2010 PMID 20466091). Patients with autism/autism spectrum disorders (ASDs). Suspected autosomal recessive condition due to close familial relations Previously negative karyotyping and/or chromosomal microarray results. Test Description This panel analyzes 2429 genes that have been associated with Autism and ID and/or disorders associated with Autism and ID. Both sequencing and deletion/duplication (CNV) analysis will be performed on the coding regions of all genes included (unless otherwise marked). All analysis is performed utilizing Next Generation Sequencing (NGS) technology. CNV analysis is designed to detect the majority of deletions and duplications of three exons or greater in size. Smaller CNV events may also be detected and reported, but additional follow-up testing is recommended if a smaller CNV is suspected. All variants are classified according to ACMG guidelines. Condition Description Autism Spectrum Disorder (ASD) refers to a group of developmental disabilities that are typically associated with challenges of varying severity in the areas of social interaction, communication, and repetitive/restricted behaviors.
    [Show full text]
  • Autocrine IFN Signaling Inducing Profibrotic Fibroblast Responses By
    Downloaded from http://www.jimmunol.org/ by guest on September 23, 2021 Inducing is online at: average * The Journal of Immunology , 11 of which you can access for free at: 2013; 191:2956-2966; Prepublished online 16 from submission to initial decision 4 weeks from acceptance to publication August 2013; doi: 10.4049/jimmunol.1300376 http://www.jimmunol.org/content/191/6/2956 A Synthetic TLR3 Ligand Mitigates Profibrotic Fibroblast Responses by Autocrine IFN Signaling Feng Fang, Kohtaro Ooka, Xiaoyong Sun, Ruchi Shah, Swati Bhattacharyya, Jun Wei and John Varga J Immunol cites 49 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2013/08/20/jimmunol.130037 6.DC1 This article http://www.jimmunol.org/content/191/6/2956.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 23, 2021. The Journal of Immunology A Synthetic TLR3 Ligand Mitigates Profibrotic Fibroblast Responses by Inducing Autocrine IFN Signaling Feng Fang,* Kohtaro Ooka,* Xiaoyong Sun,† Ruchi Shah,* Swati Bhattacharyya,* Jun Wei,* and John Varga* Activation of TLR3 by exogenous microbial ligands or endogenous injury-associated ligands leads to production of type I IFN.
    [Show full text]
  • Insulin Resistance and Diabetes Mellitus in Alzheimer's Disease
    cells Review Insulin Resistance and Diabetes Mellitus in Alzheimer’s Disease Jesús Burillo 1,2,3,† , Patricia Marqués 1,3,†, Beatriz Jiménez 1,2,3,† , Carlos González-Blanco 1,3,†, Manuel Benito 1,2,3,‡ and Carlos Guillén 1,2,3,*,‡ 1 Department of Biochemistry, Complutense University, 28040 Madrid, Spain; [email protected] (J.B.); [email protected] (P.M.); [email protected] (B.J.); [email protected] (C.G.-B.); [email protected] (M.B.) 2 Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain 3 Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain * Correspondence: [email protected] † These authors contribute equally to this review. ‡ These authors contribute equally to this review. Abstract: Type 2 diabetes mellitus is a progressive disease that is characterized by the appearance of insulin resistance. The term insulin resistance is very wide and could affect different proteins involved in insulin signaling, as well as other mechanisms. In this review, we have analyzed the main molecular mechanisms that could be involved in the connection between type 2 diabetes and neurodegeneration, in general, and more specifically with the appearance of Alzheimer’s disease. We have studied, in more detail, the different processes involved, such as inflammation, endoplasmic reticulum stress, autophagy, and mitochondrial dysfunction. Citation: Burillo, J.; Marqués, P.; Jiménez, B.; González-Blanco, C.; Keywords: insulin resistance; T3DM; mTOR; ER stress; autophagy; inflammation Benito, M.; Guillén, C. Insulin Resistance and Diabetes Mellitus in Alzheimer’s Disease. Cells 2021, 10, 1236. https://doi.org/10.3390/ 1.
    [Show full text]