Functional Characterization of Histidine Domain-Protein Tyrosine Phosphatase (HD-PTP) in Tumor Development

Total Page:16

File Type:pdf, Size:1020Kb

Functional Characterization of Histidine Domain-Protein Tyrosine Phosphatase (HD-PTP) in Tumor Development Functional characterization of histidine domain-protein tyrosine phosphatase (HD-PTP) in tumor development By Sanaz Manteghi A thesis submitted to McGill University in partial fulfillment of the requirements of the degree of Doctor of Philosophy Department of Biochemistry McGill University Montreal, QC, Canada December 2015 © Sanaz Manteghi, 2015 1 Abstract Histidine Domain containing Protein Tyrosine Phosphatase (HD-PTP/PTPN23) is part of the ESCRT (Endosomal Sorting Complex Required for Transport) complex with previously established function in cell surface receptor sorting, signaling and multi-vesicular body biogenesis. HD-PTP is encoded by the PTPN23 gene, which maps to chromosomal region 3p21.3, reported to be frequently deleted in various human cancers. However, the bona fide tumor suppressive role of HD-PTP has not been demonstrated yet. In this study we have investigated Ptpn23’s tumor suppressor function in vivo and we further assessed its role in cellular transformation and NF-κB pathway modulation in vitro. Components of the ESCRT complex have long been proposed to act as tumor suppressors. Here we show for the first time that PTPN23/HD-PTP, an ESCRT associated protein, exhibits tumor suppressor function in vivo. Ptpn23 hemizygous knockout mice are susceptible to the development of spontaneous B-cell lymphoma and lung adenocarcinoma. Moreover, B-cell lymphoma onset and dissemination were significantly potentiated by hemizygous loss of Ptpn23 in the transgenic Eµ-myc mouse model. Tumors derived from the Ptpn23+/- mice maintained HD- PTP expression and exhibited impaired apoptosis together with reduced levels of the ARF tumor suppressor. In addition, PTPN23 hemizygous loss resulted in partial cell transformation and increased migration/invasion. Furthermore, analysis of human cancer gene expression data revealed that heterozygous loss of PTPN23 is common in many human cancers, which is correlated with poor survival. This suggests that PTPN23/HD-PTP is a prominent tumor suppressor gene in human malignancies. Moreover, we identified a new function for HD-PTP in TNFα-induced canonical NF-κB activation downstream of TNFR1 receptor. We have shown that HD-PTP depleted cells have 2 increased cell surface TNFR1 and increased TNFR1 signaling complex formation which occurs due to defect in TNFR1 degradation in the context of HD-PTP deficiency. Furthermore, we have identified HD-PTP as a negative regulator of NF-κB pathway via TNFR1 receptor. We have shown that HD-PTP depleted cells display faster kinetics of NF-κB activation, increased target gene expression and inflammatory cytokine secretion upon TNFα stimulation. This suggests that HD- PTP inhibits NF-κB activation by controlling cell surface TNFR1 levels and recruitment of adaptor molecules to TNFR1 in order to attenuate the signaling cascade. In conclusion, these data suggest that PTPN23 is a haplo-insufficient tumor suppressor gene involved in tumor initiation, progression and NF-κB signaling, demonstrating for the first time the importance of an ESCRT component, HD-PTP, in tumorigenesis and immunity. Further investigation is required to determine the exact molecular mechanism of HD-PTP in tumorigenesis, inflammation and cancer. 3 Résumé La proteine Histidine Domain containing Protein Tyrosine Phosphatase (HD-PTP) fait partie du complexe ESCRT (Endosomal Sorting Complex Required for Transport), un complexe multiprotéique responsable du triage endosomal des récepteurs cellulaires de surface, ainsi que de la signalisation et de la formation des endosomes multi-vésiculaires (MVBs). La protéine HD-PTP est le produit du gène PTPN23, localisé dans la région chromosomale 3p21.3, une région fréquemment perdue dans plusieurs types de cancers humains. Néanmoins, la fonction suppresseur de tumeurs de HD-PTP n’a jamais été démontrée. Cette étude évalue la fonction suppresseur de tumeurs de HD-PTP in vivo, ainsi que le rôle de HD-PTP dans la transformation cellulaire et dans la régulation de la voie de signalisation NF-κB. Il a été précédemment suggéré que les constituants du complexe ESCRT agissent comme suppresseurs de tumeurs. Dans cette étude, nous démontrons pour la première fois la fonction suppresseur de tumeur de HD-PTP, une protéine associée avec la machinerie ESCRT. En effet, les souris hémizygotes pour Ptpn23 sont susceptibles au développement spontané de lymphomes à cellules B et d’adénocarcinomes pulmonaires. De plus, la perte d’une allèle de Ptpn23 potentialise significativement l’apparition et la dissémination de lymphomes induits dans le modèle transgénique murin exprimant l’oncogène Myc (Eμ-Myc). Les tumeurs dérivées des souris Ptpn23+/- préservent l’expression hémizygote de HD-PTP et démontrent des niveaux réduits d’apoptose et d’expression du suppresseur de tumeurs ARF. De plus, la déficience hémizygote de Ptpn23 induit la transformation partielle des cellules et augmente le phénotype pro-migratoire et pro-invasif de celles-ci. L’analyse de données d’expression génique de cancers humains révèle que la perte partielle de l’expression de PTPN23 est fréquente dans plusieurs types de tumeurs humaines et corrèle avec un mauvais pronostic. 4 Par ailleurs, nous avons identifié une nouvelle fonction de HD-PTP dans la régulation de la voie de signalisation canonique de NF-κB suite à une stimulation du récepteur TNFR1 avec TNFα. Nous avons démontré que les cellules déficientes pour HD-PTP ont des niveaux élevés de TNFR1 à la surface membranaire et une formation accrue de complexe de signalisation recruté subséquemment. Ces effets sont dus à une diminution de la dégradation de TNFR1 lorsque l’expression de HD-PTP est supprimée. Collectivement, ces effets mènent à une activation plus rapide de NF-κB et de l’expression de gènes spécifiques à cette cascade de signalisation, ainsi que l’augmentation de la sécrétion de cytokines pro-inflammatoires suite à une stimulation avec le TNFα. Nos résultats suggèrent que HD-PTP agit comme un inhibiteur de la voie de signalisation de NF-κB en modulant les niveaux d’expression de TNFR1 à la surface cellulaire et en régulant l’assemblement du complexe de signalisation afin d’atténuer la signalisation en aval. En résumé, nos résultats suggèrent fortement que Ptpn23 est un suppresseur de tumeurs haplo-insuffisant impliqué dans l’initiation de tumeurs, dans leur progression et dans la voie de signalisation NF-κB. Ces observations confirment pour la première fois le rôle suppresseur de tumeurs d’une composante de la machinerie ESCRT. Des études supplémentaires seront nécessaires afin d’élucider le mécanisme moléculaire précis de HD-PTP régulant l’inflammation et le cancer. 5 Acknowledgements I would like to thank My supervisor Dr. Arnim Pause who directed my training with continual guidance, and patience and for giving me the opportunity to work on a very challenging project. Present and former members of my research advisory committee, Dr. Jerry Pelletier, Dr. Maya Saleh, Dr. Russell Jones and Dr. Gerardo Ferbeyre for their valuable advice, critical comments and encouragement. My best colleague Dmitri Kharitidi, for being such an excellent person to work with on a daily basis during the last 5 years. My present and former labmates, Elena Malitskaya, Jin Yong Patrick Park, Luc Galarneau, Mikhail Chetcherbinine, Debora Cote, Anders Dydensborg, Yann Nouet, Ming Yan, Zahra Jalali, Mathieu Paquette, Tarika Vijayaraghavan, Sanket Khan, Elite Possik, (their contributions are listed in each chapter). Dr. Marie-Claude Gingras who initially started HD-PTP mouse model and for her help and assistance in this project. Dr. Marilène Paquet for performing pathology analysis. Dr. Regina Cencic and Dr. Francis Robert from Dr. Pelletier lab for their help with mouse experiments. Dr. Maud Marques from Dr. Michael Witcher’s lab, who kindly taught me to perform bioinformatic analysis of TCGA data. Dr. Maya Poffenberger and Eric Ma for performing IL6 ELISA assays. Dr. John R. Mills and Dr. Abba Malina for the insightful discussions on myc project. 6 Jin Yong Patrick Park, Anisa Domi, Takla Gris and Said Izreig for their help with editing of different chapters of this thesis. My family and my friends who are always there to help me through good and bad times and for their endless support, and encouraging words to keep me on track. Funding: Supported by a McGill Integrated Cancer Research Training Program (MICRTP) studentship award, a Canderel studentship award, a George G. Harris fellowship award in cancer, a McGill Biochemistry Graduate Excellence Fellowship award, and grants from the Cancer Research Society (CRS), the Canadian Cancer Society Research Institute (CCSRI: #700525 and #702500 to AP) and the McGill Collaborative Research and Development Fund. 7 Preface This thesis is written in manuscript format and resulted in one published review paper, two papers under revision and two papers in collaboration, one published and one under preparation for submission. 1. Dmitri Kharitidi*, Sanaz Manteghi* and Arnim Pause, Pseudo phosphatases: identification methods and physiological significance, Methods, S1046-2023(13)00378-2, (2013). (*Equally contributed) 2. Sanaz Manteghi, Marie-Claude Gingras, Dmitri Kharitidi, Luc Galarneau, Maud Marques, Ming Yan, Regina Cencic, Francis Robert, Marilène Paquet, Michael Witcher, Jerry Pelletier and Arnim Pause, Haploinsufficiency of the ESCRT component HD-PTP predisposes to cancer (in revision at Cell Reports) 3. Sanaz Manteghi*, Dmitri Kharitidi*, Jin Yong Patrick Park, Mikhail Chetcherbinine, Maya Poffenberger,
Recommended publications
  • The Expression Patterns and the Prognostic Roles of PTPN Family Members in Digestive Tract Cancers
    Preprint: Please note that this article has not completed peer review. The expression patterns and the prognostic roles of PTPN family members in digestive tract cancers CURRENT STATUS: UNDER REVIEW Jing Chen The First Affiliated Hospital of China Medical University Xu Zhao Liaoning Vocational College of Medicine Yuan Yuan The First Affiliated Hospital of China Medical University Jing-jing Jing The First Affiliated Hospital of China Medical University [email protected] Author ORCiD: https://orcid.org/0000-0002-9807-8089 DOI: 10.21203/rs.3.rs-19689/v1 SUBJECT AREAS Cancer Biology KEYWORDS PTPN family members, digestive tract cancers, expression, prognosis, clinical features 1 Abstract Background Non-receptor protein tyrosine phosphatases (PTPNs) are a set of enzymes involved in the tyrosyl phosphorylation. The present study intended to clarify the associations between the expression patterns of PTPN family members and the prognosis of digestive tract cancers. Method Expression profiling of PTPN family genes in digestive tract cancers were analyzed through ONCOMINE and UALCAN. Gene ontology enrichment analysis was conducted using the DAVID database. The gene–gene interaction network was performed by GeneMANIA and the protein–protein interaction (PPI) network was built using STRING portal couple with Cytoscape. Data from The Cancer Genome Atlas (TCGA) were downloaded for validation and to explore the relationship of the PTPN expression with clinicopathological parameters and survival of digestive tract cancers. Results Most PTPN family members were associated with digestive tract cancers according to Oncomine, Ualcan and TCGA data. For esophageal carcinoma (ESCA), expression of PTPN1, PTPN4 and PTPN12 were upregulated; expression of PTPN20 was associated with poor prognosis.
    [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]
  • RT² Profiler PCR Array (96-Well Format and 384-Well [4 X 96] Format)
    RT² Profiler PCR Array (96-Well Format and 384-Well [4 x 96] Format) Human Protein Phosphatases Cat. no. 330231 PAHS-045ZA For pathway expression analysis Format For use with the following real-time cyclers RT² Profiler PCR Array, Applied Biosystems® models 5700, 7000, 7300, 7500, Format A 7700, 7900HT, ViiA™ 7 (96-well block); Bio-Rad® models iCycler®, iQ™5, MyiQ™, MyiQ2; Bio-Rad/MJ Research Chromo4™; Eppendorf® Mastercycler® ep realplex models 2, 2s, 4, 4s; Stratagene® models Mx3005P®, Mx3000P®; Takara TP-800 RT² Profiler PCR Array, Applied Biosystems models 7500 (Fast block), 7900HT (Fast Format C block), StepOnePlus™, ViiA 7 (Fast block) RT² Profiler PCR Array, Bio-Rad CFX96™; Bio-Rad/MJ Research models DNA Format D Engine Opticon®, DNA Engine Opticon 2; Stratagene Mx4000® RT² Profiler PCR Array, Applied Biosystems models 7900HT (384-well block), ViiA 7 Format E (384-well block); Bio-Rad CFX384™ RT² Profiler PCR Array, Roche® LightCycler® 480 (96-well block) Format F RT² Profiler PCR Array, Roche LightCycler 480 (384-well block) Format G RT² Profiler PCR Array, Fluidigm® BioMark™ Format H Sample & Assay Technologies Description The Human Protein Phosphatases RT² Profiler PCR Array profiles the gene expression of the 84 most important and well-studied phosphatases in the mammalian genome. By reversing the phosphorylation of key regulatory proteins mediated by protein kinases, phosphatases serve as a very important complement to kinases and attenuate activated signal transduction pathways. The gene classes on this array include both receptor and non-receptor tyrosine phosphatases, catalytic subunits of the three major protein phosphatase gene families, the dual specificity phosphatases, as well as cell cycle regulatory and other protein phosphatases.
    [Show full text]
  • NCOMMS-19-37816 the Manuscript by Hay Et Al Describes the Structure An
    Reviewers' comments: Reviewer #1 (Remarks to the Author): NCOMMS-19-37816 The manuscript by Hay et al describes the structure and characterization of the PTPRU pseudophosphatase, a receptor tyrosine phosphatase that is proposed to be proto-oncogenic, although its true function is still not fully established. The biological function of this protein is the question addressed in this manuscript, which describes biochemical, structural and cell biological data unified by an effort to uncover its true function in cells. The experiments are solid and interesting, providing new intriguing possibilities about the role of this protein in biology and cancer. That being said, some of the conclusions lack the solidity one might expect from a Nature Communications paper and thus a few additional studies are proposed that are designed to strengthen the manuscript conclusions, especially regarding mechanism. Unraveling these questions are very important to the field of phosphorylation signaling. 1.Activity assays: The enzyme activity assays are solid and convincing. However, no rationale is provided as to why PTPRK was selected for the comparative studies and a sentence or two at the beginning of this paragraph would strengthen this entire section (it took a couple of looks at the figure to realize some of the data refer to PTPRK). If the PTP loop Cys is replaced by Ser, are there any phenotypes? Similarly, is it perfectly conserved across all organisms that contain PTPRU genes? This might also provide insights into the role of this protein (and confirm lack of activity is ‘fine’). 2.Structure: This section is very interesting, but a few additional experiments would strengthen the conclusions.
    [Show full text]
  • The Human Gene Connectome As a Map of Short Cuts for Morbid Allele Discovery
    The human gene connectome as a map of short cuts for morbid allele discovery Yuval Itana,1, Shen-Ying Zhanga,b, Guillaume Vogta,b, Avinash Abhyankara, Melina Hermana, Patrick Nitschkec, Dror Friedd, Lluis Quintana-Murcie, Laurent Abela,b, and Jean-Laurent Casanovaa,b,f aSt. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065; bLaboratory of Human Genetics of Infectious Diseases, Necker Branch, Paris Descartes University, Institut National de la Santé et de la Recherche Médicale U980, Necker Medical School, 75015 Paris, France; cPlateforme Bioinformatique, Université Paris Descartes, 75116 Paris, France; dDepartment of Computer Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; eUnit of Human Evolutionary Genetics, Centre National de la Recherche Scientifique, Unité de Recherche Associée 3012, Institut Pasteur, F-75015 Paris, France; and fPediatric Immunology-Hematology Unit, Necker Hospital for Sick Children, 75015 Paris, France Edited* by Bruce Beutler, University of Texas Southwestern Medical Center, Dallas, TX, and approved February 15, 2013 (received for review October 19, 2012) High-throughput genomic data reveal thousands of gene variants to detect a single mutated gene, with the other polymorphic genes per patient, and it is often difficult to determine which of these being of less interest. This goes some way to explaining why, variants underlies disease in a given individual. However, at the despite the abundance of NGS data, the discovery of disease- population level, there may be some degree of phenotypic homo- causing alleles from such data remains somewhat limited. geneity, with alterations of specific physiological pathways under- We developed the human gene connectome (HGC) to over- come this problem.
    [Show full text]
  • Live-Cell Imaging Rnai Screen Identifies PP2A–B55α and Importin-Β1 As Key Mitotic Exit Regulators in Human Cells
    LETTERS Live-cell imaging RNAi screen identifies PP2A–B55α and importin-β1 as key mitotic exit regulators in human cells Michael H. A. Schmitz1,2,3, Michael Held1,2, Veerle Janssens4, James R. A. Hutchins5, Otto Hudecz6, Elitsa Ivanova4, Jozef Goris4, Laura Trinkle-Mulcahy7, Angus I. Lamond8, Ina Poser9, Anthony A. Hyman9, Karl Mechtler5,6, Jan-Michael Peters5 and Daniel W. Gerlich1,2,10 When vertebrate cells exit mitosis various cellular structures can contribute to Cdk1 substrate dephosphorylation during vertebrate are re-organized to build functional interphase cells1. This mitotic exit, whereas Ca2+-triggered mitotic exit in cytostatic-factor- depends on Cdk1 (cyclin dependent kinase 1) inactivation arrested egg extracts depends on calcineurin12,13. Early genetic studies in and subsequent dephosphorylation of its substrates2–4. Drosophila melanogaster 14,15 and Aspergillus nidulans16 reported defects Members of the protein phosphatase 1 and 2A (PP1 and in late mitosis of PP1 and PP2A mutants. However, the assays used in PP2A) families can dephosphorylate Cdk1 substrates in these studies were not specific for mitotic exit because they scored pro- biochemical extracts during mitotic exit5,6, but how this relates metaphase arrest or anaphase chromosome bridges, which can result to postmitotic reassembly of interphase structures in intact from defects in early mitosis. cells is not known. Here, we use a live-cell imaging assay and Intracellular targeting of Ser/Thr phosphatase complexes to specific RNAi knockdown to screen a genome-wide library of protein substrates is mediated by a diverse range of regulatory and targeting phosphatases for mitotic exit functions in human cells. We subunits that associate with a small group of catalytic subunits3,4,17.
    [Show full text]
  • Phosphatases Page 1
    Phosphatases esiRNA ID Gene Name Gene Description Ensembl ID HU-05948-1 ACP1 acid phosphatase 1, soluble ENSG00000143727 HU-01870-1 ACP2 acid phosphatase 2, lysosomal ENSG00000134575 HU-05292-1 ACP5 acid phosphatase 5, tartrate resistant ENSG00000102575 HU-02655-1 ACP6 acid phosphatase 6, lysophosphatidic ENSG00000162836 HU-13465-1 ACPL2 acid phosphatase-like 2 ENSG00000155893 HU-06716-1 ACPP acid phosphatase, prostate ENSG00000014257 HU-15218-1 ACPT acid phosphatase, testicular ENSG00000142513 HU-09496-1 ACYP1 acylphosphatase 1, erythrocyte (common) type ENSG00000119640 HU-04746-1 ALPL alkaline phosphatase, liver ENSG00000162551 HU-14729-1 ALPP alkaline phosphatase, placental ENSG00000163283 HU-14729-1 ALPP alkaline phosphatase, placental ENSG00000163283 HU-14729-1 ALPPL2 alkaline phosphatase, placental-like 2 ENSG00000163286 HU-07767-1 BPGM 2,3-bisphosphoglycerate mutase ENSG00000172331 HU-06476-1 BPNT1 3'(2'), 5'-bisphosphate nucleotidase 1 ENSG00000162813 HU-09086-1 CANT1 calcium activated nucleotidase 1 ENSG00000171302 HU-03115-1 CCDC155 coiled-coil domain containing 155 ENSG00000161609 HU-09022-1 CDC14A CDC14 cell division cycle 14 homolog A (S. cerevisiae) ENSG00000079335 HU-11533-1 CDC14B CDC14 cell division cycle 14 homolog B (S. cerevisiae) ENSG00000081377 HU-06323-1 CDC25A cell division cycle 25 homolog A (S. pombe) ENSG00000164045 HU-07288-1 CDC25B cell division cycle 25 homolog B (S. pombe) ENSG00000101224 HU-06033-1 CDKN3 cyclin-dependent kinase inhibitor 3 ENSG00000100526 HU-02274-1 CTDSP1 CTD (carboxy-terminal domain,
    [Show full text]
  • Tubular P53 Regulates Multiple Genes to Mediate AKI
    BASIC RESEARCH www.jasn.org Tubular p53 Regulates Multiple Genes to Mediate AKI † † † † † Dongshan Zhang,* Yu Liu,* Qingqing Wei, Yuqing Huo, Kebin Liu, Fuyou Liu,* and † Zheng Dong* *Departments of Emergency Medicine and Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China; and †Department of Cellular Biology and Anatomy, Vascular Biology Center and Department of Biochemistry and Molecular Biology, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia ABSTRACT A pathogenic role of p53 in AKI was suggested a decade ago but remains controversial. Indeed, recent work indicates that inhibition of p53 protects against ischemic AKI in rats but exacerbates AKI in mice. One intriguing possibility is that p53 has cell type-specific roles in AKI. To determine the role of tubular p53, we generated two conditional gene knockout mouse models, in which p53 is specifically ablated from proximal tubules or other tubular segments, including distal tubules, loops of Henle, and medullary collecting ducts. Proximal tubule p53 knockout (PT-p53-KO) mice were resistant to ischemic and cisplatin nephrotoxic AKI, which was indicated by the analysis of renal function, histology, apoptosis, and inflammation. However, other tubular p53 knockout (OT-p53-KO) mice were sensitive to AKI. Mechanis- tically, AKI associated with the upregulation of several known p53 target genes, including Bax, p53- upregulated modulator of apoptosis-a, p21, and Siva, and this association was attenuated in PT-p53-KO mice. In global expression analysis, ischemic AKI induced 371 genes in wild-type kidney cortical tissues, but the induction of 31 of these genes was abrogated in PT-p53-KO tissues.
    [Show full text]
  • SND1 Mediated Downregulation of PTPN23 in HCC
    Virginia Commonwealth University VCU Scholars Compass Theses and Dissertations Graduate School 2014 SND1 mediated downregulation of PTPN23 in HCC Nidhi Jariwala Virginia Commonwealth University Follow this and additional works at: https://scholarscompass.vcu.edu/etd Part of the Genetics Commons, and the Molecular Genetics Commons © The Author Downloaded from https://scholarscompass.vcu.edu/etd/3648 This Thesis is brought to you for free and open access by the Graduate School at VCU Scholars Compass. It has been accepted for inclusion in Theses and Dissertations by an authorized administrator of VCU Scholars Compass. For more information, please contact [email protected]. SND1 Mediated Downregulation of PTPN23 in Hepatocellular Carcinoma A thesis submitted in partial fulfillment of the requirements for the degree of Master of Science Virginia Commonwealth University By NIDHI JARIWALA, MS. Department of Biotechnology, University of Mumbai, India, 2012 ADVISOR: DR. DEVANAND SARKAR, M.B.B.S., Ph.D. Associate Professor, Department of Human and Molecular Genetics Blick Scholar Associate Scientific Director, Cancer Therapeutics VCU Institute of Molecular Medicine Massey Cancer Center Virginia Commonwealth University Richmond, Virginia December, 2014 ii Acknowledgement I am grateful to my teacher, Dr. Devanand Sarkar for not just guiding me in research but inspiring me to become a refined scientist. For inculcating in me, appreciation of hard work and perseverance. He teaches by example, the value of ethics, honesty, sincerity and good deed. I feel not only fortunate but very proud to be his student. I have been blessed by stellar mentors and thank all my teachers, who play a vital role in building not just my career, but also my character.
    [Show full text]
  • EGFR Signaling from the Early Endosome
    University of Louisville ThinkIR: The University of Louisville's Institutional Repository Electronic Theses and Dissertations 8-2018 EGFR signaling from the early endosome. Julie A. Gosney University of Louisville Follow this and additional works at: https://ir.library.louisville.edu/etd Part of the Biochemistry Commons, Cancer Biology Commons, Cell Biology Commons, and the Pharmacology Commons Recommended Citation Gosney, Julie A., "EGFR signaling from the early endosome." (2018). Electronic Theses and Dissertations. Paper 3007. https://doi.org/10.18297/etd/3007 This Doctoral Dissertation is brought to you for free and open access by ThinkIR: The University of Louisville's Institutional Repository. It has been accepted for inclusion in Electronic Theses and Dissertations by an authorized administrator of ThinkIR: The University of Louisville's Institutional Repository. This title appears here courtesy of the author, who has retained all other copyrights. For more information, please contact [email protected]. EGFR SIGNALING FROM THE EARLY ENDOSOME By Julie A. Gosney B.A., Anderson University, 2012 M.S., University of Louisville, 2016 A Dissertation Submitted to the Faculty of the School of Medicine of the University of Louisville In Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy in Pharmacology and Toxicology Department of Pharmacology and Toxicology University of Louisville Louisville, KY August 2018 Copyright 2018 by Julie Gosney All Rights Reserved EGFR SIGNALING FROM THE EARLY ENDOSOME By Julie A. Gosney B.A., Anderson University, 2012 M.S., University of Louisville Dissertation Approved on June 6, 2018 By the following Dissertation Committee: ______________________________________ Brian P. Ceresa, Ph.D. ______________________________________ Joshua Hood, M.D., Ph.D.
    [Show full text]
  • Integrin-Linked Kinase Controls Renal Branching Morphogenesis Via Dual Specificity Phosphatase 8
    BASIC RESEARCH www.jasn.org Integrin-linked Kinase Controls Renal Branching Morphogenesis via Dual Specificity Phosphatase 8 †‡ Joanna Smeeton,* Priya Dhir,* Di Hu,* Meghan M. Feeney,* Lin Chen,* and † Norman D. Rosenblum* § *Program in Developmental and Stem Cell Biology, and §Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada; and †Departments of Paediatrics, and ‡Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada ABSTRACT Integrin-linked kinase (ILK) is an intracellular scaffold protein with critical cell-specific functions in the embryonic and mature mammalian kidney. Previously, we demonstrated a requirement for Ilk during ureteric branching and cell cycle regulation in collecting duct cells in vivo. Although in vitro data indicate that ILK controls p38 mitogen-activated protein kinase (p38MAPK) activity, the contribution of ILK- p38MAPK signaling to branching morphogenesis in vivo is not defined. Here, we identified genes that are regulated by Ilk in ureteric cells using a whole-genome expression analysis of whole-kidney mRNA in mice with Ilk deficiency in the ureteric cell lineage. Six genes with expression in ureteric tip cells, including Wnt11, were downregulated, whereas the expression of dual-specificity phosphatase 8 (DUSP8) was upregulated. Phosphorylation of p38MAPK was decreased in kidney tissue with Ilk deficiency, but no significant decrease in the phosphorylation of other intracellular effectors previously shown to control renal morphogenesis was observed. Pharmacologic inhibition of p38MAPK activity in murine inner med- ullary collecting duct 3 (mIMCD3) cells decreased expression of Wnt11, Krt23,andSlo4c1.DUSP8over- expression in mIMCD3 cells significantly inhibited p38MAPK activation and the expression of Wnt11 and Slo4c1. Adenovirus-mediated overexpression of DUSP8 in cultured embryonic murine kidneys decreased ureteric branching and p38MAPK activation.
    [Show full text]
  • Protein Tyrosine Phosphatases in Health and Disease Wiljan J
    REVIEW ARTICLE Protein tyrosine phosphatases in health and disease Wiljan J. A. J. Hendriks1, Ari Elson2, Sheila Harroch3, Rafael Pulido4, Andrew Stoker5 and Jeroen den Hertog6,7 1 Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands 2 Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel 3 Department of Neuroscience, Institut Pasteur, Paris, France 4 Centro de Investigacio´ nPrı´ncipe Felipe, Valencia, Spain 5 Neural Development Unit, Institute of Child Health, University College London, UK 6 Hubrecht Institute, KNAW & University Medical Center Utrecht, The Netherlands 7 Institute of Biology Leiden, Leiden University, The Netherlands Keywords Protein tyrosine phosphatases (PTPs) represent a super-family of enzymes bone morphogenesis; hereditary disease; that play essential roles in normal development and physiology. In this neuronal development; post-translational review, we will discuss the PTPs that have a causative role in hereditary modification; protein tyrosine phosphatase; diseases in humans. In addition, recent progress in the development and synaptogenesis analysis of animal models expressing mutant PTPs will be presented. The Correspondence impact of PTP signaling on health and disease will be exemplified for the J. den Hertog, Hubrecht Institute, fields of bone development, synaptogenesis and central nervous system dis- Uppsalalaan 8, 3584 CT Utrecht, eases. Collectively, research on PTPs since the late 1980’s yielded the The Netherlands cogent view that development of PTP-directed
    [Show full text]