Comparison of Predicted and Actual Consequences of Missense Mutations

Total Page:16

File Type:pdf, Size:1020Kb

Comparison of Predicted and Actual Consequences of Missense Mutations Comparison of predicted and actual consequences of PNAS PLUS missense mutations Lisa A. Miosgea,1, Matthew A. Fielda,1, Yovina Sontania, Vicky Choa,b, Simon Johnsona,b, Anna Palkovaa,b, Bhavani Balakishnanb, Rong Liangb, Yafei Zhangb, Stephen Lyonc, Bruce Beutlerc, Belinda Whittleb, Edward M. Bertramb, Anselm Endersd, Christopher C. Goodnowa,e,2,3, and T. Daniel Andrewsa,2,3 aImmunogenomics Laboratory, John Curtin School of Medical Research, Australian National University, Canberra City, ACT 2601, Australia; bAustralian Phenomics Facility, John Curtin School of Medical Research, Australian National University, Canberra City, ACT 2601, Australia; cCenter for Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390; dRamaciotti Immunisation Genomics Laboratory, John Curtin School of Medical Research, Australian National University, Canberra City, ACT 2601, Australia; and eImmunogenomics Laboratory, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia Contributed by Christopher C. Goodnow, July 9, 2015 (sent for review February 24, 2015; reviewed by Jean-Laurent Casanova and Alain Fischer) Each person’s genome sequence has thousands of missense vari- Many tools now exist that use diverse information to make in- ants. Practical interpretation of their functional significance must ferences of the functional importance of single amino acid sub- rely on computational inferences in the absence of exhaustive stitutions (3, 4). The majority of better-performing tools use a experimental measurements. Here we analyzed the efficacy of protein multiple sequence alignment and judge the importance of these inferences in 33 de novo missense mutations revealed by residues depending on their conservation across available ho- sequencing in first-generation progeny of N-ethyl-N-nitrosourea– mologous sequences. Notably, these tools are sensitive to the treated mice, involving 23 essential immune system genes. Poly- sequence choice in the input alignment (5). Some examples of Phen2, SIFT, MutationAssessor, Panther, CADD, and Condel were commonly used tools that use a sequence conservation approach, ’ used to predict each mutation s functional importance, whereas with diverse methods to calculate conservation, are SIFT (6), the actual effect was measured by breeding and testing homozy- MutationAssessor (7), MAPP (8), AlignGVGD (9), PANTHER gotes for the expected in vivo loss-of-function phenotype. Only (10), and GERP (11). Protein structural information is also in- 20% of mutations predicted to be deleterious by PolyPhen2 (and cluded in other tools to judge whether an amino acid substitution INFLAMMATION 15% by CADD) showed a discernible phenotype in individual ho- IMMUNOLOGY AND may importantly alter protein stability or catalysis, but few com- mozygotes. Half of all possible missense mutations in the same monly used tools rely just on these data alone (3). 23 immune genes were predicted to be deleterious, and most of The widely used PolyPhen2 and CADD tools (12, 13) integrate these appear to become subject to purifying selection because few persist between separate mouse substrains, rodents, or primates. a number of different information sources, including sequence- Because defects in immune genes could be phenotypically masked and structure-based features (and in the case of CADD, the re- in vivo by compensation and environment, we compared infere- sults of other tools), and use a machine learning approach to nces by the same tools with the in vitro phenotype of all 2,314 categorize variants as benign or deleterious. It is worth noting that possible missense variants in TP53; 42% of mutations predicted by inference tools that integrate diverse information, such as PolyPhen2 PolyPhen2 to be deleterious (and 45% by CADD) had little measur- able consequence for TP53-promoted transcription. We conclude Significance that for de novo or low-frequency missense mutations found by genome sequencing, half those inferred as deleterious correspond Computational tools applied to any human genome sequence to nearly neutral mutations that have little impact on the clinical identify hundreds of genetic variants predicted to disrupt the phenotype of individual cases but will nevertheless become sub- function of individual proteins as the result of a single codon ject to purifying selection. change. These tools have been trained on disease mutations and common polymorphisms but have yet to be tested against an de novo mutation | immunodeficiency | evolution | nearly neutral | cancer unbiased spectrum of random mutations arising de novo. Here we perform such a test comparing the predicted and actual ef- he genome sequence of any particular person contains ex- fects of de novo mutations in 23 genes with essential functions Ttensive protein-altering genetic variation and de novo point for normal immunity and all possible mutations in the TP53 tu- mutations (1), of which only a minority are unambiguously dele- mor suppressor gene. These results highlight an important gap terious and introduce premature stop codons or disrupt normal in our ability to relate genotype to phenotype in clinical genome mRNA splicing. When considering a person with a suspected sequencing: the inability to differentiate immediately clinically genetic illness, the first clinical question is whether any of the relevant mutations from nearly neutral mutations. mutations identified in their genome sequence involve essential genes whose disruption is known to cause a phenotype resembling Author contributions: L.A.M., M.A.F., B. Beutler, B.W., E.M.B., A.E., C.C.G., and T.D.A. designed research; L.A.M., M.A.F., Y.S., V.C., S.J., A.P., B. Balakishnan, R.L., Y.Z., S.L., B.W., that of the patient in question. The most numerous class of pro- and T.D.A. performed research; S.L., B. Beutler, and T.D.A. contributed new reagents/analytic tein-altering mutations is missense mutations, where a single co- tools; L.A.M., M.A.F., Y.S., V.C., S.J., A.P., B. Balakishnan, R.L., Y.Z., S.L., B. Beutler, B.W., E.M.B., don is altered to encode a different amino acid. On average, 2% of A.E., C.C.G., and T.D.A. analyzed data; and M.A.F., C.C.G. and T.D.A. wrote the paper. people carry a missense mutation in any given gene (2). Hence, by Reviewers: J.-L.C., The Rockefeller University; and A.F., Imagine Institute, Hôpital Necker chance, missense mutations will often be found in genes that are Enfants Malades. seemingly relevant to a person’s disease phenotype, and the next The authors declare no conflict of interest. key clinical question is whether or not these substitutions alter the Freely available online through the PNAS open access option. function of the corresponding protein. Short of mutational studies 1L.A.M. and M.A.F. contributed equally to this work. of all possible amino acid substitutions coupled with comprehen- 2C.C.G. and T.D.A. contributed equally to this work. sive functional assays, the sheer number and diversity of missense 3To whom correspondence may be addressed. Email: [email protected] or dan. mutations present in each person’s genome means that their [email protected]. functional importance must presently be addressed primarily by This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. computational inference. 1073/pnas.1511585112/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1511585112 PNAS Early Edition | 1of10 Downloaded by guest on September 25, 2021 and SNAP (14), do not necessarily have efficacy better than sim- each gene in the mouse genome, we developed a resource of mice pler tools (3). Interestingly, there is further utility in integrating the with identified mutations and corresponding exome sequence data predictions of individual methods (15), even those that are very (databases.apf.edu.au/mutations and mutagenetix.org/incidental/ similar or that already integrate wide ranges of information, po- incidental_list.cfm). First generation (G1) offspring from male tentially due to minimization of outlier effects. mice treated with ENU have a mean of 45 nonsynonymous de Functional inferences of the severity of protein disruption only novo mutations (23), although the functional effect of each mu- weakly predict disease incidence, disease severity, or clinical tation is largely unknown. Mutations induced by exposure to ENU outcome. For example, functional inferences of missense vari- are likely to have diverse functional consequences, from benign to ants in the cystic fibrosis gene, CFTR, are not well correlated severely deleterious. This diversity replicates that of spontaneous with disease incidence or severity (16). Similarly, the functional nonsynonymous de novo mutations that arise in humans at an inference of mutation severity in the tumor suppressor gene, average rate of 0.75 mutations per child (24). It is also likely to TP53, does not correlate significantly with patient clinical out- replicate the functional spectrum of inherited nonsynonymous come (17). An emerging consensus has formed that functional variants in recessive genes that occur at frequencies below 1% in inference scores lack the sensitivity and specificity for their the human population. clinical use (18–20). The human genome of any particular indi- To gain an understanding of the functional effects of these vidual likely contains many apparently unambiguous disease- phenotypically unselected point mutations, we propagated 33 associated variants (1), and these very often occur in people without mutations within 23
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]
  • CD29 Identifies IFN-Γ–Producing Human CD8+ T Cells With
    + CD29 identifies IFN-γ–producing human CD8 T cells with an increased cytotoxic potential Benoît P. Nicoleta,b, Aurélie Guislaina,b, Floris P. J. van Alphenc, Raquel Gomez-Eerlandd, Ton N. M. Schumacherd, Maartje van den Biggelaarc,e, and Monika C. Wolkersa,b,1 aDepartment of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; bLandsteiner Laboratory, Oncode Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; cDepartment of Research Facilities, Sanquin Research, 1066 CX Amsterdam, The Netherlands; dDivision of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; and eDepartment of Molecular and Cellular Haemostasis, Sanquin Research, 1066 CX Amsterdam, The Netherlands Edited by Anjana Rao, La Jolla Institute for Allergy and Immunology, La Jolla, CA, and approved February 12, 2020 (received for review August 12, 2019) Cytotoxic CD8+ T cells can effectively kill target cells by producing therefore developed a protocol that allowed for efficient iso- cytokines, chemokines, and granzymes. Expression of these effector lation of RNA and protein from fluorescence-activated cell molecules is however highly divergent, and tools that identify and sorting (FACS)-sorted fixed T cells after intracellular cytokine + preselect CD8 T cells with a cytotoxic expression profile are lacking. staining. With this top-down approach, we performed an un- + Human CD8 T cells can be divided into IFN-γ– and IL-2–producing biased RNA-sequencing (RNA-seq) and mass spectrometry cells. Unbiased transcriptomics and proteomics analysis on cytokine- γ– – + + (MS) analyses on IFN- and IL-2 producing primary human producing fixed CD8 T cells revealed that IL-2 cells produce helper + + + CD8 Tcells.
    [Show full text]
  • Inflammatory Cytokine Signalling by Protein Tyrosine Phosphatases in Pancreatic Β-Cells
    59 4 W J STANLEY and others PTPN1 and PTPN6 modulate 59: 4 325–337 Research cytokine signalling in β-cells Differential regulation of pro- inflammatory cytokine signalling by protein tyrosine phosphatases in pancreatic β-cells William J Stanley1,2, Prerak M Trivedi1,2, Andrew P Sutherland1, Helen E Thomas1,2 and Esteban N Gurzov1,2,3 Correspondence should be addressed 1 St. Vincent’s Institute of Medical Research, Melbourne, Australia to E N Gurzov 2 Department of Medicine, St. Vincent’s Hospital, The University of Melbourne, Melbourne, Australia Email 3 ULB Center for Diabetes Research, Universite Libre de Bruxelles (ULB), Brussels, Belgium esteban.gurzov@unimelb. edu.au Abstract Type 1 diabetes (T1D) is characterized by the destruction of insulin-producing β-cells Key Words by immune cells in the pancreas. Pro-inflammatory including TNF-α, IFN-γ and IL-1β f pancreatic β-cells are released in the islet during the autoimmune assault and signal in β-cells through f protein tyrosine phosphorylation cascades, resulting in pro-apoptotic gene expression and eventually phosphatases β-cell death. Protein tyrosine phosphatases (PTPs) are a family of enzymes that regulate f PTPN1 phosphorylative signalling and are associated with the development of T1D. Here, we f PTPN6 observed expression of PTPN6 and PTPN1 in human islets and islets from non-obese f cytokines diabetic (NOD) mice. To clarify the role of these PTPs in β-cells/islets, we took advantage f inflammation Journal of Molecular Endocrinology of CRISPR/Cas9 technology and pharmacological approaches to inactivate both proteins. We identify PTPN6 as a negative regulator of TNF-α-induced β-cell death, through JNK- dependent BCL-2 protein degradation.
    [Show full text]
  • Changes in Peripheral and Local Tumor Immunity After Neoadjuvant Chemotherapy Reshape Clinical Outcomes in Patients with Breast Cancer Margaret L
    Published OnlineFirst August 21, 2020; DOI: 10.1158/1078-0432.CCR-19-3685 CLINICAL CANCER RESEARCH | TRANSLATIONAL CANCER MECHANISMS AND THERAPY Changes in Peripheral and Local Tumor Immunity after Neoadjuvant Chemotherapy Reshape Clinical Outcomes in Patients with Breast Cancer Margaret L. Axelrod1, Mellissa J. Nixon1, Paula I. Gonzalez-Ericsson2, Riley E. Bergman1, Mark A. Pilkinton3, Wyatt J. McDonnell3, Violeta Sanchez1,2, Susan R. Opalenik1, Sherene Loi4, Jing Zhou5, Sean Mackay5, Brent N. Rexer1, Vandana G. Abramson1, Valerie M. Jansen1, Simon Mallal3, Joshua Donaldson1, Sara M. Tolaney6, Ian E. Krop6, Ana C. Garrido-Castro6, Jonathan D. Marotti7,8, Kevin Shee9, Todd. W. Miller8,9, Melinda E. Sanders2,10, Ingrid A. Mayer1,2, Roberto Salgado4,11, and Justin M. Balko1,2 ABSTRACT ◥ Purpose: The recent approval of anti-programmed death-ligand Results: In non-TNBC, no change in expression of any single 1 immunotherapy in combination with nab-paclitaxel for meta- gene was associated with RFS or OS, while in TNBC upregulation of static triple-negative breast cancer (TNBC) highlights the need to multiple immune-related genes and gene sets were associated with understand the role of chemotherapy in modulating the tumor improved long-term outcome. High cytotoxic T-cell signatures immune microenvironment (TIME). present in the peripheral blood of patients with breast cancer at Experimental Design: We examined immune-related gene surgery were associated with persistent disease and recurrence, expression patterns before and after neoadjuvant chemotherapy suggesting active antitumor immunity that may indicate ongoing (NAC) in a series of 83 breast tumors, including 44 TNBCs, from disease burden. patients with residual disease (RD).
    [Show full text]
  • The Regulatory Roles of Phosphatases in Cancer
    Oncogene (2014) 33, 939–953 & 2014 Macmillan Publishers Limited All rights reserved 0950-9232/14 www.nature.com/onc REVIEW The regulatory roles of phosphatases in cancer J Stebbing1, LC Lit1, H Zhang, RS Darrington, O Melaiu, B Rudraraju and G Giamas The relevance of potentially reversible post-translational modifications required for controlling cellular processes in cancer is one of the most thriving arenas of cellular and molecular biology. Any alteration in the balanced equilibrium between kinases and phosphatases may result in development and progression of various diseases, including different types of cancer, though phosphatases are relatively under-studied. Loss of phosphatases such as PTEN (phosphatase and tensin homologue deleted on chromosome 10), a known tumour suppressor, across tumour types lends credence to the development of phosphatidylinositol 3--kinase inhibitors alongside the use of phosphatase expression as a biomarker, though phase 3 trial data are lacking. In this review, we give an updated report on phosphatase dysregulation linked to organ-specific malignancies. Oncogene (2014) 33, 939–953; doi:10.1038/onc.2013.80; published online 18 March 2013 Keywords: cancer; phosphatases; solid tumours GASTROINTESTINAL MALIGNANCIES abs in sera were significantly associated with poor survival in Oesophageal cancer advanced ESCC, suggesting that they may have a clinical utility in Loss of PTEN (phosphatase and tensin homologue deleted on ESCC screening and diagnosis.5 chromosome 10) expression in oesophageal cancer is frequent, Cao et al.6 investigated the role of protein tyrosine phosphatase, among other gene alterations characterizing this disease. Zhou non-receptor type 12 (PTPN12) in ESCC and showed that PTPN12 et al.1 found that overexpression of PTEN suppresses growth and protein expression is higher in normal para-cancerous tissues than induces apoptosis in oesophageal cancer cell lines, through in 20 ESCC tissues.
    [Show full text]
  • Human Lectins, Their Carbohydrate Affinities and Where to Find Them
    biomolecules Review Human Lectins, Their Carbohydrate Affinities and Where to Review HumanFind Them Lectins, Their Carbohydrate Affinities and Where to FindCláudia ThemD. Raposo 1,*, André B. Canelas 2 and M. Teresa Barros 1 1, 2 1 Cláudia D. Raposo * , Andr1 é LAQVB. Canelas‐Requimte,and Department M. Teresa of Chemistry, Barros NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829‐516 Caparica, Portugal; [email protected] 12 GlanbiaLAQV-Requimte,‐AgriChemWhey, Department Lisheen of Chemistry, Mine, Killoran, NOVA Moyne, School E41 of ScienceR622 Co. and Tipperary, Technology, Ireland; canelas‐ [email protected] NOVA de Lisboa, 2829-516 Caparica, Portugal; [email protected] 2* Correspondence:Glanbia-AgriChemWhey, [email protected]; Lisheen Mine, Tel.: Killoran, +351‐212948550 Moyne, E41 R622 Tipperary, Ireland; [email protected] * Correspondence: [email protected]; Tel.: +351-212948550 Abstract: Lectins are a class of proteins responsible for several biological roles such as cell‐cell in‐ Abstract:teractions,Lectins signaling are pathways, a class of and proteins several responsible innate immune for several responses biological against roles pathogens. such as Since cell-cell lec‐ interactions,tins are able signalingto bind to pathways, carbohydrates, and several they can innate be a immuneviable target responses for targeted against drug pathogens. delivery Since sys‐ lectinstems. In are fact, able several to bind lectins to carbohydrates, were approved they by canFood be and a viable Drug targetAdministration for targeted for drugthat purpose. delivery systems.Information In fact, about several specific lectins carbohydrate were approved recognition by Food by andlectin Drug receptors Administration was gathered for that herein, purpose. plus Informationthe specific organs about specific where those carbohydrate lectins can recognition be found by within lectin the receptors human was body.
    [Show full text]
  • University of California Santa Cruz Sample
    UNIVERSITY OF CALIFORNIA SANTA CRUZ SAMPLE-SPECIFIC CANCER PATHWAY ANALYSIS USING PARADIGM A dissertation submitted in partial satisfaction of the requirements for the degree of DOCTOR OF PHILOSOPHY in BIOMOLECULAR ENGINEERING AND BIOINFORMATICS by Stephen C. Benz June 2012 The Dissertation of Stephen C. Benz is approved: Professor David Haussler, Chair Professor Joshua Stuart Professor Nader Pourmand Dean Tyrus Miller Vice Provost and Dean of Graduate Studies Copyright c by Stephen C. Benz 2012 Table of Contents List of Figures v List of Tables xi Abstract xii Dedication xiv Acknowledgments xv 1 Introduction 1 1.1 Identifying Genomic Alterations . 2 1.2 Pathway Analysis . 5 2 Methods to Integrate Cancer Genomics Data 10 2.1 UCSC Cancer Genomics Browser . 11 2.2 BioIntegrator . 16 3 Pathway Analysis Using PARADIGM 20 3.1 Method . 21 3.2 Comparisons . 26 3.2.1 Distinguishing True Networks From Decoys . 27 3.2.2 Tumor versus Normal - Pathways associated with Ovarian Cancer 29 3.2.3 Differentially Regulated Pathways in ER+ve vs ER-ve breast can- cers . 36 3.2.4 Therapy response prediction using pathways (Platinum Free In- terval in Ovarian Cancer) . 38 3.3 Unsupervised Stratification of Cancer Patients by Pathway Activities . 42 4 SuperPathway - A Global Pathway Model for Cancer 51 4.1 SuperPathway in Ovarian Cancer . 55 4.2 SuperPathway in Breast Cancer . 61 iii 4.2.1 Chin-Naderi Cohort . 61 4.2.2 TCGA Breast Cancer . 63 4.3 Cross-Cancer SuperPathway . 67 5 Pathway Analysis of Drug Effects 74 5.1 SuperPathway on Breast Cell Lines .
    [Show full text]
  • Supplementary Data
    Progressive Disease Signature Upregulated probes with progressive disease U133Plus2 ID Gene Symbol Gene Name 239673_at NR3C2 nuclear receptor subfamily 3, group C, member 2 228994_at CCDC24 coiled-coil domain containing 24 1562245_a_at ZNF578 zinc finger protein 578 234224_at PTPRG protein tyrosine phosphatase, receptor type, G 219173_at NA NA 218613_at PSD3 pleckstrin and Sec7 domain containing 3 236167_at TNS3 tensin 3 1562244_at ZNF578 zinc finger protein 578 221909_at RNFT2 ring finger protein, transmembrane 2 1552732_at ABRA actin-binding Rho activating protein 59375_at MYO15B myosin XVB pseudogene 203633_at CPT1A carnitine palmitoyltransferase 1A (liver) 1563120_at NA NA 1560098_at AKR1C2 aldo-keto reductase family 1, member C2 (dihydrodiol dehydrogenase 2; bile acid binding pro 238576_at NA NA 202283_at SERPINF1 serpin peptidase inhibitor, clade F (alpha-2 antiplasmin, pigment epithelium derived factor), m 214248_s_at TRIM2 tripartite motif-containing 2 204766_s_at NUDT1 nudix (nucleoside diphosphate linked moiety X)-type motif 1 242308_at MCOLN3 mucolipin 3 1569154_a_at NA NA 228171_s_at PLEKHG4 pleckstrin homology domain containing, family G (with RhoGef domain) member 4 1552587_at CNBD1 cyclic nucleotide binding domain containing 1 220705_s_at ADAMTS7 ADAM metallopeptidase with thrombospondin type 1 motif, 7 232332_at RP13-347D8.3 KIAA1210 protein 1553618_at TRIM43 tripartite motif-containing 43 209369_at ANXA3 annexin A3 243143_at FAM24A family with sequence similarity 24, member A 234742_at SIRPG signal-regulatory protein gamma
    [Show full text]
  • Activation of Islet Inflammation by Cytokine Signalling in Pancreatic Β
    Activation of Islet Inflammation by Cytokine Signalling in Pancreatic b-cells Understanding the Role of Protein Tyrosine Phosphatases William James Stanley ORCID: 0000-0001-8001-2359 Student Number: 515467 Submitted in Total Fulfilment of The Degree of Doctor of Philosophy March 2020 St. Vincent’s Institute of Medical Research Department of Medicine (St. Vincent’s Hospital) The University of Melbourne Abstract: Type 1 diabetes is characterised by the autoimmune destruction of insulin producing β- cells in the islets of Langerhans of the pancreas. Immune cells release pro-inflammatory cytokines such as interferon-g (IFN-g), tumour necrosis factor-a (TNF-a) and interleukin- 1b (IL-1b) into the islet microenvironment which activate phosphorylation cascades and gene expression in b-cells that increase their susceptibility to autoimmune attack and destruction. Protein tyrosine phosphatases (PTPs) regulate phosphorylation based signalling pathways and have previously been shown to negatively regulate IFN-g induced cell death of β-cells in vitro. We previously showed that during immune infiltration to the islet PTPs, including PTPN1 and PTPN6, are rendered catalytically inactive through oxidation resulting in loss of signal regulation. The overall aim of this thesis is to observe if antioxidant treatment can reduce autoimmune development in the NOD/Lt mouse through reduction of oxidised PTPs and dissect the role of PTPN1 and PTPN6 in the regulation of cytotoxic signalling events in the NIT-1 β-cell line and isolated NODPI islets in vitro. Chapter 3 studies the effect of the mitochondrial targeted antioxidant mito-TEMPO on insulitis and diabetes development in the NOD/Lt mouse. Delivery of mito-TEMPO through drinking water reduced levels of oxidised PTPs in the pancreas of NOD/Lt mice but had no effect on the development of insulitis, activity or number of CD8+ and CD4+ T- cells in the periphery in NOD/Lt mice or diabetes development in a diabetes transfer model.
    [Show full text]
  • T Cells Deficient in the Tyrosine Phosphatase SHP-1 Resist Suppression by Regulatory T Cells
    T Cells Deficient in the Tyrosine Phosphatase SHP-1 Resist Suppression by Regulatory T Cells This information is current as Emily R. Mercadante and Ulrike M. Lorenz of September 29, 2021. J Immunol published online 26 May 2017 http://www.jimmunol.org/content/early/2017/05/26/jimmun ol.1602171 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2017/05/26/jimmunol.160217 Material 1.DCSupplemental Why The JI? Submit online. http://www.jimmunol.org/ • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average by guest on September 29, 2021 Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2017 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published May 26, 2017, doi:10.4049/jimmunol.1602171 The Journal of Immunology T Cells Deficient in the Tyrosine Phosphatase SHP-1 Resist Suppression by Regulatory T Cells Emily R. Mercadante and Ulrike M. Lorenz The balance between activation of T cells and their suppression by regulatory T cells (Tregs) is dysregulated in autoimmune diseases and cancer. Autoimmune diseases feature T cells that are resistant to suppression by Tregs, whereas in cancer, T cells are unable to mount antitumor responses due to the Treg-enriched suppressive microenvironment.
    [Show full text]
  • Prognostic and Therapeutic Significance of Phosphorylated
    Han et al. Blood Cancer Journal (2018) 8:110 DOI 10.1038/s41408-018-0138-8 Blood Cancer Journal ARTICLE Open Access Prognostic and therapeutic significance of phosphorylated STAT3 and protein tyrosine phosphatase-6 in peripheral-T cell lymphoma Jing Jing Han1,MeganO’byrne2,MaryJ.Stenson1, Matthew J. Maurer 2, Linda E. Wellik1,AndrewL.Feldman 3, Ellen D. McPhail3,ThomasE.Witzig1 and Mamta Gupta1,4 Abstract Peripheral T cell lymphomas (PTCL) is a heterogenous group of non-Hodgkin lymphoma and many patients remain refractory to the frontline therapy. Identifying new prognostic markers and treatment is an unmet need in PTCL. We analyzed phospho-STAT3 (pSTAT3) expression in a cohort of 169 PTCL tumors and show overall 38% positivity with varied distribution among PTCL subtypes with 27% (16/59) in PTCL-NOS; 29% (11/38) in AITL, 57% (13/28) in ALK- negative ALCL, and 93% in ALK-pos ALCL (14/15), respectively. Correlative analysis indicated an adverse correlation between pSTAT3 and overall survival (OS). PTPN6, a tyrosine phosphatase and potential negative regulator of STAT3 activity, was suppressed in 62% of PTCL-NOS, 42% of AITL, 60% ALK-neg ALCL, and 86% of ALK-pos ALCL. Loss of PTPN6 combined with pSTAT3 positivity predicted an infwere considered significantferior OS in PTCL cases. In vitro treatment of TCL lines with azacytidine (aza), a DNA methyltransferase inhibitor (DNMTi), restored PTPN6 expression 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; and decreased pSTAT3. Combining DNMTi with JAK3 inhibitor resulted in synergistic antitumor activity in SUDHL1 cell line. Overall, our results suggest that PTPN6 and activated STAT3 can be developed as prognostic markers, and the combination of DNMTi and JAK3 inhibitors as a novel treatment for patients with PTCL subtypes.
    [Show full text]
  • Alteration of the MYB Locus in MYST3-Linked Cases
    Leukemia (2009) 23, 85–94 & 2009 Macmillan Publishers Limited All rights reserved 0887-6924/09 $32.00 www.nature.com/leu ORIGINAL ARTICLE Genome profiling of acute myelomonocytic leukemia: alteration of the MYB locus in MYST3-linked cases A Murati1,13, C Gervais2,13, N Carbuccia1,13, P Finetti1, N Cervera1, J Ade´laı¨de1, S Struski2, E Lippert3, F Mugneret4, I Tigaud5, D Penther6, C Bastard6, B Poppe7, F Speleman7, L Baranger8, I Luquet9, P Cornillet-Lefebvre9, N Nadal10, F Nguyen-Khac11, CPe´rot12, S Olschwang1, F Bertucci1, M Chaffanet1, M Lessard2, M-J Mozziconacci1 and D Birnbaum1 on behalf of the Groupe Francophone de Cytoge´ne´tique He´matologique (GFCH) 1De´partement d’Oncologie Mole´culaire, Centre de Recherche en Cance´rologie de Marseille, Institut Paoli-Calmettes, Marseille, France; 2Laboratoire d’He´matologie, CHU de Hautepierre, Strasbourg, France; 3Laboratoire d’He´matologie, Hoˆpital Cardiologique-Haut Le´veˆque, Bordeaux, France; 4Laboratoire de Cytoge´ne´tique, CHU du Bocage, Dijon, France; 5Laboratoire d’He´matologie, CHU Lyon Sud, Lyon, France; 6De´partement de Ge´ne´tique, Centre Henri Becquerel, Rouen, France; 7Centre de Ge´ne´tique Me´dicale, Ghent, Belgique; 8Laboratoire de Ge´ne´tique, CHU d’Angers, Angers, France; 9Service de Ge´ne´tique, Hoˆpital Maison Blanche, Reims, France; 10Laboratoire d’He´matologie, Hoˆpital Nord, Saint-Etienne, France; 11Laboratoire de Cytoge´ne´tique He´matologique, Hoˆpital Pitie´-Salpeˆtrie`re, Paris, France and 12Laboratoire de Cytoge´ne´tique Onco-He´matologique, Hoˆpital Saint-Antoine, Paris, France The t(8;16)(p11;p13) is a rare translocation involved in de novo NCOA3 at 20q12.6 MYST3-linked acute myeloid leukemias and therapy-related myelomonocytic and monocytic acute (AMLs) share specific features, such as frequent extramedullary leukemia.
    [Show full text]