Alterations in the Rap1 Signaling Pathway Are Common in Human Gliomas

Total Page:16

File Type:pdf, Size:1020Kb

Alterations in the Rap1 Signaling Pathway Are Common in Human Gliomas Oncogene (1997) 15, 1611 ± 1616 1997 Stockton Press All rights reserved 0950 ± 9232/97 $12.00 SHORT REPORT Alterations in the rap1 signaling pathway are common in human gliomas David H Gutmann1, Susan Saporito-Irwin1, Jerey E DeClue2, Ralf Wienecke2 and Abhijit Guha3 1Department of Neurology, Washington University School of Medicine, Box 8111, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA; 2Laboratory of Cellular Oncology, Building 36, Room 1D-32, National Cancer Institute, Bethesda, Maryland 20892, USA; 3Lunenfeld Research Institute, Mount Sinai Hospital, Division of Neurosurgery, The Toronto Hospital, University of Toronto, 2-415 McLaughlin; 399 Bathurst, Toronto, Ontario M5T 2S8, Canada Several inherited predisposition to cancer syndromes are To this end, the TSC2 gene product, tuberin, functions associated with the development of nervous system as a negative regulator of rap1. Loss of tuberin tumors. Tuberous sclerosis complex (TSC) is an expression has recently been demonstrated by our autosomal dominant disorder in which aected indivi- laboratory in one third of sporadic gliomas, suggesting duals are at risk for developing astrocytomas. One of the that alterations in the rap1 signaling pathway may be genes responsible for this disorder is TSC2, located on associated with the development of human astrocyto- chromosome 16p, and encoding a 180 kDa protein mas (Wienecke et al., in press). In this study, we test (tuberin) that functions in part as a negative regulator the hypothesis that alterations in the rap1 signaling of rap1. Previous studies from our laboratory demon- pathway, either due to rap1 overexpression or reduced strated that 30% of sporadic astrocytomas have reduced levels of the rap1 negative regulator, tuberin, are or absent tuberin expression. In addition to loss of frequently associated with gliomas compared to other tuberin in sporadic astrocytomas, aberrant rap1 mediated tumors of the nervous system. signaling may also result from overexpression of rap1. In Tuberin has been demonstrated to suppress cell this study, we test the hypothesis that alterations in the growth and this activity resides in the C-terminus of rap1 signaling pathway are frequently observed in certain tuberin where the domain that functions as a GTPase subsets of gliomas compared to other tumors of the activating protein (GAP) for rap1 is located (Jin et al., nervous system. Analysis of sporadic astrocytomas and 1996). Biochemical studies demonstrated that this GAP ependymomas demonstrated either increased rap1 or activity is speci®c for rap1 and not other related small reduced/absent tuberin protein expression in 50 ± 60% of GTPase proteins, including rap2, H-ras and rho dierent cohorts of these gliomas, compared to 30 ± 33% (Wienecke et al., 1995). Loss of tuberin expression of sporadic schwannomas and meningiomas and none of would therefore be predicted to lead to elevated rap1 eight oligodendrocyte tumors. These results suggest that activity in these tumors. At present, there is no alterations in the rap1 signaling pathway are important biochemical method available to determine rap1 in the development of certain sporadic human gliomas. activity in cells to directly test this hypothesis. In addition to loss of tuberin expression, another Keywords: astrocytomas; tumor suppressor gene; mechanism for elevated rap1 signaling might result tuberous sclerosis complex 2; tuberin from increased rap1 expression. To this end, amplifica- tion of a segment of chromosome 12q containing the rap1B gene has been detected in many dierent tumor Studies on the molecular genetic events associated with types, including astrocytomas (Cheng et al., 1995; He the development of human malignant astrocytic tumors et al., 1994, 1995; Khatib et al., 1993; Reifenberger et (the most common subtype of glioma and primary al., 1996). In these tumors, there is ampli®cation of the human brain cancer) have demonstrated alterations in 12q chromosomal region variably containing the the p21-ras signaling pathway. Ampli®cation of the CDK4/SAS, MDM2, GLI, AM2R, GADD153, IFNG epidermal growth factor receptor (EGF-R) has been and RAP1B genes. detected in a high proportion of the most malignant To determine whether alterations in the rap1 astrocytoma or glioblastoma multiforme (GBM; Olson signaling pathway are observed in human astrocyto- et al., 1995), contributing to increased activation of mas, Western blot analysis was performed on twenty p21-ras. Previously, we demonstrated elevated levels of adult astrocytomas (Figure 1a). Tumor samples were p21-ras activity in astrocytoma cell lines as well as obtained from patients undergoing surgery in the fresh GBM surgical specimens (Gutmann et al., 1996; Division of Neurosurgery at the University of Toronto Guha et al., manuscript submitted). Inhibition of p21- or Washington University School of Medicine. All ras activity using either a dominant inhibitory p21-ras tumors were obtained in accordance with the respective mutant or farsenyltransferase inhibitors resulted in institutions' Institutional Review Board Human Studies reduced astrocytoma cell proliferation in vitro and Protocols and classi®ed according to the World Health tumor growth in vivo. Organization (WHO) classi®cation of tumors of the In addition to the p21-ras signaling pathway, central nervous system. For control purposes, normal activation of other small GTPase proteins may also cortex tissue was obtained from patients who underwent result in increased cell proliferation in astrocytomas. cerebral resection because of contusion. Mouse neocortical astrocyte cultures were generated as previously described (Hewett et al., 1995). The tumor Correspondence: DH Gutmann Received 14 March 1997; revised 28 May 1997; accepted samples were coded in order to conduct the experiments 28 May 1997 in a blind fashion. Brie¯y, specimens were minced and Tuberin and rap1 in nervous system tumors DH Gutmann et al 1612 lysed with the use of a Dounce homogenizer in NP40 lysis buer (50 mM Tris, pH 7.4; 150 mM NaCl; 0.5% NP-40, 1 mM DTT) and protease inhibitors while maintained on ice (Wienecke et al., in press). The a protein content was determined using a modi®ed * * * * Bradford reagent (BIORAD) and 75 micrograms of brain 317 352 379 397 410 mA 5295 52395 JPA 4295 WU47 each sample was subjected to 18% SDS polyacrylamide gel electrophoresis. The proteins were electroblotted — tuberin onto Immobilon-P membranes and the blots were incubated with the appropriate primary antibodies and developed with horseradish peroxidase-based enhanced chemiluminescence detection (ECL; Amersham). Reduced or absent tuberin expression was detected in — rap1 nine of twenty tumors relative to normal human brain or mouse astrocyte culture controls. For these studies, reduced or absent tuberin expression was de®ned as less * * than 10% of control tissue (brain or primary cultured mouse neocortical astrocytes) as determined by scan- * * * * * brain 794 805 806 808 813 820 827 834 858 859 ning densitometry. Equal loading was veri®ed using anti-actin polyclonal antibodies (data not shown; see — tuberin Figure 3b). Similar analysis of an additional 18 adult astrocytomas demonstrated reduced or absent tuberin expression in ®ve tumors. In total, reduced tuberin expression was observed in a total of 14 of 38 astrocytomas (37%). Increased rap1 expression was observed in ®ve of the 20 astrocytomas shown in Figure — rap1 1. In this study, increased rap1 expression was de®ned as greater than tenfold increased expression over control tissues (brain or primary astrocytes) as determined by scanning densitometry. The ®ve astro- * * * cytomas overexpressing rap1 had normal levels of tuberin. Similar analysis of an additional 18 adult astrocytomas demonstrated increased rap1 expression b in ®ve tumors, all of which had normal levels of tuberin brain 315 405 553 606 686 749 845 924 expression. In total, increased rap1 expression was observed in a total of 10 of 38 astrocytomas (26%). Therefore, 24 of 38 (63%) sporadic astrocytomas — tuberin demonstrated potential alterations in the rap1 signaling pathway. Consistent with the hypothesis that alterations in the rap1 signaling pathway result from either an increase in rap1 expression or a loss of tuberin expression, we observed overexpression of rap1 only in gliomas that maintained tuberin expression. There were no clinical dierences in terms of age, gender, — rap1 location of tumor and survival between this cohort of malignant astrocytomas compared to other astrocyto- mas, or between the group with decreased tuberin Figure 1 Tuberin and rap1 expression in sporadic human expression compared to those overexpressing rap1. astrocytic tumors. (a) Western blotting of protein lysates from 20 To determine if alterations in rap1 were unique to sporadic astrocytomas with the rabbit polyclonal tuberin C20 astrocytomas, we examined ependymomas and oligo- antibody (Santa Cruz Biotechnology; 1 : 1000 dilution) and mouse monoclonal rap1 antibody (Transduction Laboratories; 1 : 500 dendrogliomas (both less prevalent subtypes of human dilution) demonstrated reduced or absent tuberin expression in gliomas) as well as other non-glial nervous system nine tumors (317, 352, 5295, 4295, 813, 820, 834, 858 and 859) and tumors. In six spinal ependymomas, tuberin expression increased rap1 expression in an additional ®ve tumors (397, 410, was reduced in two tumors, while rap1 expression was 805, 806 and 827). Brain and m.A. correspond to human normal increased in an additional tumor, for a total
Recommended publications
  • Supplementary Materials
    Supplementary Materials COMPARATIVE ANALYSIS OF THE TRANSCRIPTOME, PROTEOME AND miRNA PROFILE OF KUPFFER CELLS AND MONOCYTES Andrey Elchaninov1,3*, Anastasiya Lokhonina1,3, Maria Nikitina2, Polina Vishnyakova1,3, Andrey Makarov1, Irina Arutyunyan1, Anastasiya Poltavets1, Evgeniya Kananykhina2, Sergey Kovalchuk4, Evgeny Karpulevich5,6, Galina Bolshakova2, Gennady Sukhikh1, Timur Fatkhudinov2,3 1 Laboratory of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia 2 Laboratory of Growth and Development, Scientific Research Institute of Human Morphology, Moscow, Russia 3 Histology Department, Medical Institute, Peoples' Friendship University of Russia, Moscow, Russia 4 Laboratory of Bioinformatic methods for Combinatorial Chemistry and Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia 5 Information Systems Department, Ivannikov Institute for System Programming of the Russian Academy of Sciences, Moscow, Russia 6 Genome Engineering Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia Figure S1. Flow cytometry analysis of unsorted blood sample. Representative forward, side scattering and histogram are shown. The proportions of negative cells were determined in relation to the isotype controls. The percentages of positive cells are indicated. The blue curve corresponds to the isotype control. Figure S2. Flow cytometry analysis of unsorted liver stromal cells. Representative forward, side scattering and histogram are shown. The proportions of negative cells were determined in relation to the isotype controls. The percentages of positive cells are indicated. The blue curve corresponds to the isotype control. Figure S3. MiRNAs expression analysis in monocytes and Kupffer cells. Full-length of heatmaps are presented.
    [Show full text]
  • Rap1 Acts Via Multiple Mechanisms to Position Canoe/Afadin and Adherens Junctions and Mediate Apical-Basal Polarity Establishment
    bioRxiv preprint doi: https://doi.org/10.1101/170977; this version posted July 31, 2017. 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. Rap1 acts via multiple mechanisms to position Canoe/Afadin and adherens junctions and mediate apical-basal polarity establishment Teresa T. Bonello1, Kia Z. Perez-Vale2, Kaelyn D. Sumigray3, and Mark Peifer1,2,3* 1 Department of Biology, University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC 27599-3280, USA 2 Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA 3 Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Running Title Active Rap1 positions Canoe and AJs 6950 words * To whom correspondence should be addressed Email: [email protected] Phone: (919) 962-2272 Abbreviations used: α-cat, alpha-catenin; β-cat, beta-catenin; AJ, adherens junction; Arm, Armadillo; Baz, BazooKa; CA, constitutively active; Cno, Canoe; DE-cad, Drosophila E-cadherin; Dzy, Dizzy; GAP, GTPase activating protein; GDP, guanosine diphosphate; GEF, guanine nucleotide exchange factor; GFP, green fluorescent protein; GTP, guanosine triphosphate; IF, immunofluorescence; MIP, maximum intensity projection; RA, Ras-associated; RFP, red fluorescent protein; SAJ, spot adherens junction; shRNA, short hairpin RNA; TCJ, tricellular junction; WT, wildtype 1 bioRxiv preprint doi: https://doi.org/10.1101/170977; this version posted July 31, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved.
    [Show full text]
  • Rap1 Regulates Hepatic Stellate Cell Migration Through the Modulation of Rhoa Activity in Response to TGF‑Β1
    INTERNATIONAL JOURNAL OF MOleCular meDICine 44: 491-502, 2019 Rap1 regulates hepatic stellate cell migration through the modulation of RhoA activity in response to TGF‑β1 MI-YOUNG MOON1, HEE-JUN KIM2, MO-JONG KIM2, SUNHO UHM1, JI‑WON PARK1, KI-TAE SUK3, JAE‑BONG PARK4, DONG-JUN KIM3 and SUNG-EUN KIM1 1Department of Internal Medicine, Hallym University Sacred Heart Hospital, College of Medicine, Hallym University, Anyang, Gyeonggi 14068; 2Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi 14066; 3Department of Internal Medicine, Hallym University Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, Gangwon 24253; 4Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea Received November 1, 2018; Accepted May 28, 2019 DOI: 10.3892/ijmm.2019.4215 Abstract. Although the migration of hepatic stellate cells activation of RhoA in TGF‑β1-stimulated HSC‑T6 cells. These (HSCs) is important for hepatic fibrosis, the regulation of this findings suggest that TGF‑β1 regulates Rap1, resulting in the migration is poorly understood. Notably, transforming growth suppression of RhoA, activation of and formation of F‑actin factor (TGF)-β1 induces monocyte migration to sites of injury during the migration of HSCs. or inflammation during the early phase, but inhibits cell migra- tion during the late phase. In the present study, the role of Introduction transforming protein RhoA signaling in TGF-β1-induced HSC migration was investigated. TGF‑β1 was found to increase Hepatic fibrosis is characterized by the excessive deposition the protein and mRNA levels of smooth muscle actin and of extracellular matrix (ECM) mediated by activated hepatic collagen type I in HSC‑T6 cells.
    [Show full text]
  • Rap1a and Rap1b Ras-Family Proteins Are Prominently Expressed in the Nucleus of Squamous Carcinomas: Nuclear Translocation of GTP-Bound Active Form
    Oncogene (2003) 22, 6243–6256 & 2003 Nature Publishing Group All rights reserved 0950-9232/03 $25.00 www.nature.com/onc Rap1A and rap1B ras-family proteins are prominently expressed in the nucleus of squamous carcinomas: nuclear translocation of GTP-bound active form Raj S Mitra1,4, Zhaocheng Zhang1,4, Bradley S Henson1, David M Kurnit2, Thomas ECarey 1,3, Nisha J D’Silva*,1 1Department of Oral Medicine, Pathology and Oncology, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA; 2Departments of Pediatrics and Human Genetics, University of Michigan Medical School, Ann Arbor, MI 481091, USA; 3Department of Otolaryngology, Laboratory of Head and Neck Cancer Biology, The University of Michigan Medical School and the Comprehensive Cancer Center, Ann Arbor, MI 48109-0506, USA We recently showed that rap1 regulates growth and exclusively in eukaryotes (Takai et al., 2001). Those proliferation in normal keratinocytes, which provoked us SMGs that have been identified (currently 4100) have to investigate its expression and regulation in malignant homology to ras, and belong to one of the five cells. Rap1 is variably expressed in whole cell lysates of subfamilies namely ras, rho, rab, sar1/arf and ran squamous cell carcinoma (SCC) cell lines. Immunoblot (Takai et al., 2001). These ras-like proteins shuttle analysis of nuclear and cytosolic fractions and immuno- between inactive GDP- and active GTP-bound states. histochemistry revealed that in addition to cytoplasmic Ras subfamily proteins, including ras and rap1, are key expression, SCC cells also exhibit prominent punctate players in receptor-linked signaling pathways that rap1 expression in the nucleus.
    [Show full text]
  • Distinct Actions of Rab3 and Rab27 Gtpases on Late Stages of Exocytosis of Insulin
    © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd doi:10.1111/tra.12182 Distinct Actions of Rab3 and Rab27 GTPases on Late Stages of Exocytosis of Insulin Victor A. Cazares1,2, Arasakumar Subramani1, Johnny J. Saldate1,2, Widmann Hoerauf1,2 and Edward L. Stuenkel1,2,∗ 1Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA 2Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA ∗Corresponding author: Edward L. Stuenkel, [email protected] Abstract Rab GTPases associated with insulin-containing secretory granules (SGs) readily releasable pool (RRP). By comparison, nucleotide cycling of Rab3 are key in targeting, docking and assembly of molecular complexes GTPases, but not of Rab27A, is essential for a kinetically rapid filling of governing pancreatic β-cell exocytosis. Four Rab3 isoforms along with the RRP with SGs. Aside from these distinct functions, Rab3 and Rab27A Rab27A are associated with insulin granules, yet elucidation of the GTPases demonstrate considerable functional overlap in building the distinct roles of these Rab families on exocytosis remains unclear. To readily releasable granule pool. Hence, while Rab3 and Rab27A coop- define specific actions of these Rab families we employ Rab3GAP erate to generate release-ready SGs in β-cells, they also direct unique and/or EPI64A GTPase-activating protein overexpression in β-cells from kinetic and functional properties of the exocytotic pathway. wild-type or Ashen mice to selectively transit the entire Rab3 fam- Keywords exocytosis, GTPase, insulin secretion, membrane fusion, Rab ily or Rab27A to a GDP-bound state. Ashen mice carry a sponta- proteins neous mutation that eliminates Rab27A expression.
    [Show full text]
  • Site-Specific Processing of Ras and Rap1 Switch I by a MARTX Toxin
    ARTICLE Received 8 Jan 2015 | Accepted 1 May 2015 | Published 8 Jun 2015 DOI: 10.1038/ncomms8396 OPEN Site-specific processing of Ras and Rap1 Switch I by a MARTX toxin effector domain Irena Antic1,*, Marco Biancucci1,*, Yueming Zhu2, David R. Gius2 & Karla J.F. Satchell1 Ras (Rat sarcoma) protein is a central regulator of cell growth and proliferation. Mutations in the RAS gene are known to occur in human cancers and have been shown to contribute to carcinogenesis. In this study, we show that the multifunctional-autoprocessing repeats-in- toxin (MARTX) toxin-effector domain DUF5Vv from Vibrio vulnificus to be a site-specific endopeptidase that cleaves within the Switch 1 region of Ras and Rap1. DUF5Vv processing of Ras, which occurs both biochemically and in mammalian cell culture, inactivates ERK1/2, thereby inhibiting cell proliferation. The ability to cleave Ras and Rap1 is shared by DUF5Vv homologues found in other bacteria. In addition, DUF5Vv can cleave all Ras isoforms and KRas with mutations commonly implicated in malignancies. Therefore, we speculate that this new family of Ras/Rap1-specific endopeptidases (RRSPs) has potential to inactivate both wild-type and mutant Ras proteins expressed in malignancies. 1 Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Ward 6-225, Chicago, Illinois 60611, USA. 2 Department of Radiation Oncology and Pharmacology, Feinberg School of Medicine, Northwestern University, 303 East Superior Avenue, Lurie 3-119, Chicago, Illinois 60611, USA. * These authors contributed equally to this work. Correspondence and requests for materials should be addressedto K.J.F.S.
    [Show full text]
  • Small Gtpases of the Ras and Rho Families Switch On/Off Signaling
    International Journal of Molecular Sciences Review Small GTPases of the Ras and Rho Families Switch on/off Signaling Pathways in Neurodegenerative Diseases Alazne Arrazola Sastre 1,2, Miriam Luque Montoro 1, Patricia Gálvez-Martín 3,4 , Hadriano M Lacerda 5, Alejandro Lucia 6,7, Francisco Llavero 1,6,* and José Luis Zugaza 1,2,8,* 1 Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; [email protected] (A.A.S.); [email protected] (M.L.M.) 2 Department of Genetics, Physical Anthropology, and Animal Physiology, Faculty of Science and Technology, UPV/EHU, 48940 Leioa, Spain 3 Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, 180041 Granada, Spain; [email protected] 4 R&D Human Health, Bioibérica S.A.U., 08950 Barcelona, Spain 5 Three R Labs, Science Park of the UPV/EHU, 48940 Leioa, Spain; [email protected] 6 Faculty of Sport Science, European University of Madrid, 28670 Madrid, Spain; [email protected] 7 Research Institute of the Hospital 12 de Octubre (i+12), 28041 Madrid, Spain 8 IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain * Correspondence: [email protected] (F.L.); [email protected] (J.L.Z.) Received: 25 July 2020; Accepted: 29 August 2020; Published: 31 August 2020 Abstract: Small guanosine triphosphatases (GTPases) of the Ras superfamily are key regulators of many key cellular events such as proliferation, differentiation, cell cycle regulation, migration, or apoptosis. To control these biological responses, GTPases activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in some small GTPases also guanine nucleotide dissociation inhibitors (GDIs).
    [Show full text]
  • Feedback Regulation of G Protein-Coupled Receptor Signaling
    G Model YSCDB-1903; No. of Pages 10 ARTICLE IN PRESS Seminars in Cell & Developmental Biology xxx (2016) xxx–xxx Contents lists available at ScienceDirect Seminars in Cell & Developmental Biology j ournal homepage: www.elsevier.com/locate/semcdb Review Feedback regulation of G protein-coupled receptor signaling by GRKs and arrestins a b a,∗ Joseph B. Black , Richard T. Premont , Yehia Daaka a Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, United States b Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States a r a t b i c s t l e i n f o r a c t Article history: GPCRs are ubiquitous in mammalian cells and present intricate mechanisms for cellular signaling Received 23 October 2015 and communication. Mechanistically, GPCR signaling was identified to occur vectorially through het- Accepted 19 December 2015 erotrimeric G proteins that are negatively regulated by GRK and arrestin effectors. Emerging evidence Available online xxx highlights additional roles for GRK and Arrestin partners, and establishes the existence of interconnected feedback pathways that collectively define GPCR signaling. GPCRs influence cellular dynamics and can Keywords: mediate pathologic development, such as cancer and cardiovascular remolding. Hence, a better under- GPCR standing of their overall signal regulation is of great translational interest and research continues to G protein Arrestin exploit the pharmacologic potential for modulating their activity. GRK © 2016 Elsevier Ltd. All rights reserved. Signal transduction Biased signaling Contents 1. Introduction . 00 2. 7TMR/GPCR signaling overview . 00 2.1. Diversity . 00 2.2. Structure and classification .
    [Show full text]
  • The Bitter Taste Receptor Tas2r14 Is Expressed in Ovarian Cancer and Mediates Apoptotic Signalling
    THE BITTER TASTE RECEPTOR TAS2R14 IS EXPRESSED IN OVARIAN CANCER AND MEDIATES APOPTOTIC SIGNALLING by Louis T. P. Martin Submitted in partial fulfilment of the requirements for the degree of Master of Science at Dalhousie University Halifax, Nova Scotia June 2017 © Copyright by Louis T. P. Martin, 2017 DEDICATION PAGE To my grandparents, Christina, Frank, Brenda and Bernie, and my parents, Angela and Tom – for teaching me the value of hard work. ii TABLE OF CONTENTS LIST OF TABLES ............................................................................................................. vi LIST OF FIGURES .......................................................................................................... vii ABSTRACT ....................................................................................................................... ix LIST OF ABBREVIATIONS AND SYMBOLS USED .................................................... x ACKNOWLEDGEMENTS .............................................................................................. xii CHAPTER 1 INTRODUCTION ........................................................................................ 1 1.1 G-PROTEIN COUPLED RECEPTORS ................................................................ 1 1.2 GPCR CLASSES .................................................................................................... 4 1.3 GPCR SIGNALING THROUGH G PROTEINS ................................................... 6 1.4 BITTER TASTE RECEPTORS (TAS2RS) ...........................................................
    [Show full text]
  • Substrate Recognition of MARTX Ras/Rap1 Specific Endopeptidase
    Substrate Recognition of MARTX Ras/Rap1-Specific Endopeptidase The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters. Citation Biancucci, Marco et al. “Substrate Recognition of MARTX Ras/Rap1- Specific Endopeptidase.” Biochemistry 56, 21 (May 2017): 2747–2757 © 2017 American Chemical Society As Published http://dx.doi.org/10.1021/ACS.BIOCHEM.7B00246 Publisher American Chemical Society (ACS) Version Author's final manuscript Citable link http://hdl.handle.net/1721.1/116350 Terms of Use Article is made available in accordance with the publisher's policy and may be subject to US copyright law. Please refer to the publisher's site for terms of use. HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Biochemistry Manuscript Author . Author manuscript; Manuscript Author available in PMC 2018 May 30. Published in final edited form as: Biochemistry. 2017 May 30; 56(21): 2747–2757. doi:10.1021/acs.biochem.7b00246. Substrate recognition of MARTX Ras/Rap1 specific endopeptidase Marco Biancucci1, Amy E. Rabideau2,†, Zeyu Lu2, Alex R. Loftis2, Bradley L. Pentelute2, and Karla J. F. Satchell1,* 1Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA 2Department of Chemistry, Massachusetts Institute of Technology, Boston, MA 02139 USA Abstract Ras/Rap1-specific endopeptidase (RRSP) is a cytotoxic effector domain of Multifunctional- autoprocessing repeats-in-toxins (MARTX) toxin of highly virulent strains of Vibrio vulnificus. RRSP blocks RAS-MAPK kinase signaling by cleaving Ras and Rap1 within the Switch I region between Y32 and D33. Although the RRSP processing site is highly conserved among small GTPases, only Ras and Rap1 have been identified as proteolytic substrates.
    [Show full text]
  • Elucidating the Role of EPAC-Rap1 Signaling Pathway in the Blood
    Elucidating the Role of EPAC-Rap1 Signaling Pathway in the Blood-Retinal Barrier By Carla Jhoana Ramos A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Cellular and Molecular Biology) in The University of Michigan 2017 Doctoral Committee: Professor David A. Antonetti, Chair Professor Christin Carter-Su Associate Professor Philip J. Gage Professor Ben Margolis Assistant Professor Ann L. Miller Image of exchange factor directly activated by cAMP (EPAC), Ras-related protein (Rap1) and Zonula occludens-1 (ZO-1) immunofluorescent staining in bovine retinal endothelial cells, obtained by confocal microscopy. Carla Jhoana Ramos [email protected] ORCID iD: 0000-0001-5041-6758 © Carla Jhoana Ramos 2017 DEDICATION This dissertation is dedicated to my mother Victoria de Jesus Larios de Ramos, my father Jose Carlos Ramos, and my younger brother Hilmar Ramos. My parents migrated from El Salvador to the USA in search of better opportunities and dedicated their lives to providing us with a good education. They have instilled in me determination, perseverance and strength. Finally, my experience observing my mother cope with more than 26 years of diabetes and 10 years of mild diabetic retinopathy led me into diabetes research at the University of Michigan. ii ACKNOWLEDGEMENTS The completion of this dissertation has been made possible by the support from my community of mentors, friends and family. I would like to first thank my mentor David Antonetti, who gave me the opportunity to train in his laboratory under his guidance during these last 6 years. Additionally, I would like to thank my thesis committee: Ben Margolis, Christin Carter-Su, Philip Gage, and Ann Miller whose suggestions, questions, and scientific discussions helped improve the quality of this thesis and for their constant support and encouragement.
    [Show full text]
  • Variations in Microrna-25 Expression Influence the Severity of Diabetic
    BASIC RESEARCH www.jasn.org Variations in MicroRNA-25 Expression Influence the Severity of Diabetic Kidney Disease † † † Yunshuang Liu,* Hongzhi Li,* Jieting Liu,* Pengfei Han, Xuefeng Li, He Bai,* Chunlei Zhang,* Xuelian Sun,* Yanjie Teng,* Yufei Zhang,* Xiaohuan Yuan,* Yanhui Chu,* and Binghai Zhao* *Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Medical Research Center, Heilongjiang, People’s Republic of China; and †Clinical Laboratory of Hong Qi Hospital, Mudanjiang Medical University, Heilongjiang, People’s Republic of China ABSTRACT Diabetic nephropathy is characterized by persistent albuminuria, progressive decline in GFR, and second- ary hypertension. MicroRNAs are dysregulated in diabetic nephropathy, but identification of the specific microRNAs involved remains incomplete. Here, we show that the peripheral blood from patients with diabetes and the kidneys of animals with type 1 or 2 diabetes have low levels of microRNA-25 (miR-25) compared with those of their nondiabetic counterparts. Furthermore, treatment with high glucose decreased the expression of miR-25 in cultured kidney cells. In db/db mice, systemic administration of an miR-25 agomir repressed glomerular fibrosis and reduced high BP. Notably, knockdown of miR-25 in normal mice by systemic administration of an miR-25 antagomir resulted in increased proteinuria, extracellular matrix accumulation, podocyte foot process effacement, and hypertension with renin-angiotensin system activation. However, excessive miR-25 did not cause kidney dysfunction in wild-type mice. RNA sequencing showed the alteration of miR-25 target genes in antagomir-treated mice, including the Ras-related gene CDC42. In vitro,cotrans- fection with the miR-25 antagomir repressed luciferase activity from a reporter construct containing the CDC42 39 untranslated region.
    [Show full text]