New Secreted Toxins and Immunity Proteins Encoded Within the Type VI Secretion System Gene Cluster of Serratia Marcescens

Total Page:16

File Type:pdf, Size:1020Kb

New Secreted Toxins and Immunity Proteins Encoded Within the Type VI Secretion System Gene Cluster of Serratia Marcescens Molecular Microbiology (2012) 86(4), 921–936 ᭿ doi:10.1111/mmi.12028 First published online 27 September 2012 New secreted toxins and immunity proteins encoded within the Type VI secretion system gene cluster of Serratia marcescens Grant English,1† Katharina Trunk,1† into how pathogens utilize antibacterial T6SSs to Vincenzo A. Rao,2 Velupillai Srikannathasan,2 overcome competitors and succeed in polymicrobial William N. Hunter2 and Sarah J. Coulthurst1* niches. 1Division of Molecular Microbiology, College of Life Sciences, University of Dundee, Dundee, UK. 2Division of Biological Chemistry and Drug Discovery, Introduction College of Life Sciences, University of Dundee, Dundee, Protein secretion systems and their substrates are central UK. to bacterial virulence and interaction with other organisms (Gerlach and Hensel, 2007). Six different secretion Summary systems (Types I–VI) are used by Gram-negative bacteria to transport specific proteins to the exterior of the bacterial Protein secretion systems are critical to bacterial viru- cell or further inject them into target cells. The most lence and interactions with other organisms. The Type recently described of these is the Type VI secretion VI secretion system (T6SS) is found in many bacterial system (T6SS) (Filloux et al., 2008). T6SSs are complex species and is used to target either eukaryotic cells or multi-protein assemblies that span both bacterial mem- competitor bacteria. However, T6SS-secreted proteins branes and inject effector proteins directly from the bac- have proven surprisingly elusive. Here, we identified terial cytoplasm into target cells (Bonemann et al., 2010; two secreted substrates of the antibacterial T6SS from Cascales and Cambillau, 2012). T6SSs are encoded by the opportunistic human pathogen, Serratia marces- large, variable gene clusters that contain 13 ‘core’ essen- cens. Ssp1 and Ssp2, both encoded within the T6SS tial components, believed to make up the basic secretion gene cluster, were confirmed as antibacterial toxins apparatus. Two core proteins, Hcp and VgrG, form the delivered by the T6SS. Four related proteins encoded extracellular part of the secretion machinery and depend around the Ssp proteins (‘Rap’ proteins) included two on a functional T6SS apparatus for their movement to the specifically conferring self-resistance (‘immunity’) outside of the bacterial cell (indeed, the presence of Hcp against T6SS-dependent Ssp1 or Ssp2 toxicity. Bio- in the secreted fraction has provided a useful assay for chemical characterization revealed specific, tight basic T6SS assembly and activity; Pukatzki et al., 2009). binding between cognate Ssp–Rap pairs, forming Hcp and VgrG most likely form a needle-like membrane- complexes of 2:2 stoichiometry. The atomic structures puncturing device related to the bacteriophage tail spike; of two Rap proteins were solved, revealing a novel this structure is believed to be pushed to the outside of the helical fold, dependent on a structural disulphide secreting cell and likely into target cells upon contraction bond, a structural feature consistent with their func- of a tail sheath-like structure (Leiman et al., 2009; Bone- tional localization. Homologues of the Serratia Ssp mann et al., 2010; Basler et al., 2012). T6SSs occur in and Rap proteins are found encoded together within many pathogenic bacteria and are implicated in virulence other T6SS gene clusters, thus they represent founder in important pathogens, including Burkholderia mallei, members of new families of T6SS-secreted and Burkholderia pseudomallei, Burkholderia cenocepacia, cognate immunity proteins. We suggest that Ssp pro- Vibrio cholerae, Aeromonas hydrophila, Edwardsiella teins are the original substrates of the S. marcescens tarda and Pseudomonas aeruginosa (Zheng and Leung, T6SS, before horizontal acquisition of other T6SS- 2007; Cascales, 2008; Jani and Cotter, 2010; de Pace secreted toxins. Molecular insight has been provided et al., 2010; Burtnick et al., 2011; Rosales-Reyes et al., 2012). In several cases, the action of such ‘anti- Accepted 3 September, 2012. *For correspondence. E-mail s.j. eukaryotic’ T6SSs appears to result in disruption of the [email protected]; Tel. (+44) 1382 386208; Fax (+44) 1382 388216. †These authors contributed equally and are listed actin cytoskeleton (Pukatzki et al., 2007; Aubert et al., alphabetically. 2008; Suarez et al., 2010). Exciting recent work has dem- © 2012 Blackwell Publishing Ltd 922 G. English et al. ᭿ onstrated that some T6SSs are used to target other bac- toxins. We have also identified and characterized the Rap teria, efficiently killing or inhibiting competitors. This has proteins, which include the cognate immunity proteins to been reported for T6SSs in P. aeruginosa, Burkholderia these toxins. Biochemical analyses demonstrated a tight thailandensis, V. cholerae and Serratia marcescens and specific interaction between secreted and immunity (Hood et al., 2010; MacIntyre et al., 2010; Schwarz et al., proteins. These secreted toxins and immunity proteins 2010; Murdoch et al., 2011). The discovery that certain represent two new protein families, co-occurring within T6SSs may be ‘antibacterial’ rather than, or in addition to, T6SS gene clusters of many other organisms. Addition- ‘anti-eukaryotic’ is highly relevant to the competitive ally, determination of high-resolution crystal structures of fitness and success of pathogens, particularly within pol- two members of the Rap protein family revealed that this ymicrobial infection sites. Such a system could provide family possesses a previously undescribed protein fold the pathogen with a large competitive advantage against that is dependent on formation of a disulphide bond. other bacteria in the host or the environment, enabling it to proliferate and mount a successful infection. Identifying the proteins secreted by T6SSs is a priority, Results as they will be the ‘effectors’ that directly act on target The T6SS gene cluster harbours self-resistance eukaryotic or bacterial cells. A special case of T6-secreted determinants and candidate secreted effectors effector is the class of ‘evolved’ VgrG proteins found in a minority of T6SSs which have extra C-terminal effector The T6SS gene cluster of S. marcescens Db10, domains, e.g. the actin cross-linking domain of V. chol- SMA2244–2281, contains 38 genes, including many with erae VgrG1 that is translocated into mammalian cells no known function (Murdoch et al., 2011). We speculated (Pukatzki et al., 2007; Jani and Cotter, 2010). However, that encoded within this cluster might be T6-secreted excluding the structural components VgrG and Hcp, very effectors and/or self-resistance determinants, the latter few ‘true’ T6SS-secreted proteins have been confirmed so preventing the T6SS-expressing cell from harming itself or far. Best characterized are three effector proteins, anti- being harmed by its isogenic (sibling) neighbours. In order bacterial toxins named Tse1–3, secreted by the antibac- to determine whether the cluster did indeed contain self- terial HSI-1 T6SS of P. aeruginosa. Tse1 and 3 are resistance determinants (such as specific immunity pro- peptidoglycan hydrolases that attack the cell wall of target teins analogous to the Tsi proteins), we generated a bacteria (Hood et al., 2010; Russell et al., 2011). Tse2 is mutant lacking the entire T6SS gene cluster (DT6SS) and active in the cytoplasm of target cells, where it efficiently examined whether it was fully resistant to the T6SS of the induces quiescence (Li et al., 2012). All three have adja- wild type strain. Co-culture of two target strains, wild type cently encoded cognate ‘immunity’ proteins (Tsi1–Tsi3) Db10 and the DT6SS mutant, each with a wild type and a which protect the secreting cell from harming itself or DclpV attacker, showed that the DT6SS mutant had lost being harmed by its sibling neighbours (Hood et al., 2010; resistance to T6SS-mediated inhibition or killing by the Russell et al., 2011). Significantly, obvious homologues of wild type strain (Fig. 1A, left). Recovery of DT6SS was the Tse and Tsi proteins are not detectable outside of decreased 100-fold when it was co-cultured with the wild P. aeruginosa (Hood et al., 2010). A recent report has also type strain, compared with when the wild type strain was identified a number of candidate T6SS substrates in co-cultured with itself. This effect was dependent on a B. thailandensis, one of which was confirmed as a new functional T6SS in the attacker as there was no loss of T6-secreted peptidoglycan amidase (Russell et al., 2012). DT6SS when it was co-cultured with a DclpV mutant. The Opportunistic Gram-negative bacteria cause a large ATPase ClpV is one of the core, structural components of proportion of problematic and antibiotic-resistant hospital- the T6SS and we have shown previously that it is essen- acquired infections. Enterobacteria (especially extended- tial for Hcp secretion and T6-mediated antibacterial killing spectrum b-lactamase producing isolates) are among the activity of S. marcescens Db10 (Murdoch et al., 2011). To leading culprits, including S. marcescens (Choi et al., simplify the analysis of multiple mutants, we defined the 2007; Lockhart et al., 2007). We previously reported that ‘resistance index’ of a strain as the difference between S. marcescens Db10 possesses a T6SS with potent anti- recovery when co-cultured with the wild type strain and bacterial activity (Murdoch et al., 2011). How this activity recovery when co-cultured with the DclpV mutant, specifi- is mediated,
Recommended publications
  • Deregulated Gene Expression Pathways in Myelodysplastic Syndrome Hematopoietic Stem Cells
    Leukemia (2010) 24, 756–764 & 2010 Macmillan Publishers Limited All rights reserved 0887-6924/10 $32.00 www.nature.com/leu ORIGINAL ARTICLE Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells A Pellagatti1, M Cazzola2, A Giagounidis3, J Perry1, L Malcovati2, MG Della Porta2,MJa¨dersten4, S Killick5, A Verma6, CJ Norbury7, E Hellstro¨m-Lindberg4, JS Wainscoat1 and J Boultwood1 1LRF Molecular Haematology Unit, NDCLS, John Radcliffe Hospital, Oxford, UK; 2Department of Hematology Oncology, University of Pavia Medical School, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; 3Medizinische Klinik II, St Johannes Hospital, Duisburg, Germany; 4Division of Hematology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden; 5Department of Haematology, Royal Bournemouth Hospital, Bournemouth, UK; 6Albert Einstein College of Medicine, Bronx, NY, USA and 7Sir William Dunn School of Pathology, University of Oxford, Oxford, UK To gain insight into the molecular pathogenesis of the the World Health Organization.6,7 Patients with refractory myelodysplastic syndromes (MDS), we performed global gene anemia (RA) with or without ringed sideroblasts, according to expression profiling and pathway analysis on the hemato- poietic stem cells (HSC) of 183 MDS patients as compared with the the French–American–British classification, were subdivided HSC of 17 healthy controls. The most significantly deregulated based on the presence or absence of multilineage dysplasia. In pathways in MDS include interferon signaling, thrombopoietin addition, patients with RA with excess blasts (RAEB) were signaling and the Wnt pathways. Among the most signifi- subdivided into two categories, RAEB1 and RAEB2, based on the cantly deregulated gene pathways in early MDS are immuno- percentage of bone marrow blasts.
    [Show full text]
  • Molecular Associations and Clinical Significance of Raps In
    ORIGINAL RESEARCH published: 21 June 2021 doi: 10.3389/fmolb.2021.677979 Molecular Associations and Clinical Significance of RAPs in Hepatocellular Carcinoma Sarita Kumari 1†, Mohit Arora 2†, Jay Singh 1, Lokesh K. Kadian 2, Rajni Yadav 3, Shyam S. Chauhan 2* and Anita Chopra 1* 1Laboratory Oncology Unit, Dr. BRA-IRCH, All India Institute of Medical Sciences, New Delhi, India, 2Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India, 3Department of Pathology, All India Institute of Medical Sciences, New Delhi, India Hepatocellular carcinoma (HCC) is an aggressive gastrointestinal malignancy with a high rate of mortality. Multiple studies have individually recognized members of RAP gene family as critical regulators of tumor progression in several cancers, including hepatocellular carcinoma. These studies suffer numerous limitations including a small sample size and lack of analysis of various clinicopathological and molecular Edited by: Veronica Aran, features. In the current study, we utilized authoritative multi-omics databases to Instituto Estadual do Cérebro Paulo determine the association of RAP gene family expression and detailed molecular and Niemeyer (IECPN), Brazil clinicopathological features in hepatocellular carcinoma (HCC). All five RAP genes Reviewed by: were observed to harbor dysregulated expression in HCC compared to normal liver Pooja Panwalkar, Weill Cornell Medicine, United States tissues. RAP2A exhibited strongest ability to differentiate tumors from the normal Jasminka Omerovic, tissues. RAP2A expression was associated with progressive tumor grade, TP53 and University of Split, Croatia CTNNB1 mutation status. Additionally, RAP2A expression was associated with the *Correspondence: Anita Chopra alteration of its copy numbers and DNA methylation. RAP2A also emerged as an [email protected] independent marker for patient prognosis.
    [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]
  • DLL1- and DLL4-Mediated Notch Signaling Is Essential for Adult Pancreatic Islet
    Page 1 of 41 Diabetes DLL1- and DLL4-mediated Notch signaling is essential for adult pancreatic islet homeostasis (running title –Role of Delta ligands in adult pancreas) Marina Rubey1,2,6*, Nirav Florian Chhabra1,2*, Daniel Gradinger1,2,7, Adrián Sanz-Moreno1, Heiko Lickert2,4,5, Gerhard K. H. Przemeck1,2, Martin Hrabě de Angelis1,2,3** 1 Helmholtz Zentrum München, Institute of Experimental Genetics and German Mouse Clinic, Neuherberg, Germany 2 German Center for Diabetes Research (DZD), Neuherberg, Germany 3 Chair of Experimental Genetics, Centre of Life and Food Sciences, Weihenstephan, Technische Universität München, Freising, Germany 4 Helmholtz Zentrum München, Institute of Diabetes and Regeneration Research and Institute of Stem Cell Research, Neuherberg, Germany 5 Technische Universität München, Medical Faculty, Munich, Germany 6 Present address Marina Rubey: WMC Healthcare GmbH, Munich, Germany 7 Present address Daniel Gradinger: PSI CRO AG, Munich, Germany *These authors contributed equally **Corresponding author: Prof. Dr. Martin Hrabě de Angelis, Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics, Ingolstädter Landstr.1, 85764 Neuherberg, Germany. Phone: +49-89-3187-3502. Fax: +49- 89-3187-3500. E-mail address: [email protected] Word count – 4088 / Figures – 7 Diabetes Publish Ahead of Print, published online February 6, 2020 Diabetes Page 2 of 41 Abstract Genes of the Notch signaling pathway are expressed in different cell types and organs at different time points during embryonic development and adulthood. The Notch ligand Delta- like 1 (DLL1) controls the decision between endocrine and exocrine fates of multipotent progenitors in the developing pancreas, and loss of Dll1 leads to premature endocrine differentiation.
    [Show full text]
  • Development of Novel Drugs Targeting Chaperones of Oncogenic K-Ras
    Farid Ahmad Siddiqui Farid Ahmad Siddiqui // Development of Novel Drugs Development of Novel Drugs Targeting Chaperones of Oncogenic K-Ras of Oncogenic Chaperones K-Ras Drugs Targeting of Novel Development Targeting Chaperones of Oncogenic K-Ras // 2021 9 789521 240317 ISBN 978-952-12-4031-7 Development of Novel Drugs Targeting Chaperones of Oncogenic K-Ras Farid Ahmad Siddiqui Cell Biology Faculty of Science and Engineering, Åbo Akademi University Turku Bioscience Centre University of Turku & Åbo Akademi University Turku, Finland, 2021 From the Turku Bioscience Centre, University of Turku and Åbo Akademi University, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland Supervised by Prof. Daniel Abankwa, PhD Department of Life Sciences and Medicine University of Luxembourg, Belval campus Luxembourg Reviewed by Prof. Olli Mikael Carpen, PhD Faculty of Medicine University of Helsinki Helsinki, Finland and Prof. Klaus Elenius, PhD Faculty of Medicine University of Turku Turku, Finland Opponent Prof. Krishnaraj Rajalingam, PhD Head, Cell Biology Unit, UMC-Mainz, Germany Author’s address Turku Bioscience Centre Åbo Akademi University Tykistökatu 6 20520 Turku Finland Email: [email protected] ISBN 978-952-12-4031-7 (printed) ISBN 978-952-12-4032-4 (digital) Painosalama Oy, Turku, Finland 2021 TABLE OF CONTENTS ABSTRACT .......................................................................................................... 6 ABSTRAKT (Swedish Abstract) .........................................................................
    [Show full text]
  • Gene Section Short Communication
    Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Short Communication RAP1A (RAP1A, member of RAS oncogene family) Jean de Gunzburg Laboratoire de Signalisation Intracellulaire et Oncogenèse INSERM U-528 Institut Curie Section de Recherche 26, rue d'Ulm, 75248 Paris Cedex 05, France (Jd) Published in Atlas Database: May 2001 Online updated version : http://AtlasGeneticsOncology.org/Genes/RAP1AID272.html DOI: 10.4267/2042/37748 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2001 Atlas of Genetics and Cytogenetics in Oncology and Haematology two isoforms, Rap1A and Rap1B that share 95% Identity identity and are encoded by two different genes. Rap1 HGNC (Hugo): RAP1A proteins share 50% identity with Ras proteins, Location: 1p13.3 including the regions involved in GDP/GTP binding (hence Rap1A has very similar biochemical properties to Ras), C-terminal CAAX domain leading to prenylation (geranylgeranylation in the case of Rap1A), and effector region identical to that of Ras proteins causing Ras and Rap1 to share some potential effectors. Probe(s) - Courtesy Mariano Rocchi, Resources for Molecular Expression Cytogenetics. Ubiquitous ; higher in brain and hemopo•etic tissues. DNA/RNA Localisation Description Rap1 is bound to membranes. In many cell types, it is found in a perinuclear compartment overlapping the 6 coding exons covering 18095 bp on chromosome 1. Golgi. Rap1 proteins (A and B) are phosphorylated near the C-ter by cAMP-dependent protein kinase. This Protein results in translocation of part of the Rap1 pool to the Description cytosol.
    [Show full text]
  • Role and Regulation of the P53-Homolog P73 in the Transformation of Normal Human Fibroblasts
    Role and regulation of the p53-homolog p73 in the transformation of normal human fibroblasts Dissertation zur Erlangung des naturwissenschaftlichen Doktorgrades der Bayerischen Julius-Maximilians-Universität Würzburg vorgelegt von Lars Hofmann aus Aschaffenburg Würzburg 2007 Eingereicht am Mitglieder der Promotionskommission: Vorsitzender: Prof. Dr. Dr. Martin J. Müller Gutachter: Prof. Dr. Michael P. Schön Gutachter : Prof. Dr. Georg Krohne Tag des Promotionskolloquiums: Doktorurkunde ausgehändigt am Erklärung Hiermit erkläre ich, dass ich die vorliegende Arbeit selbständig angefertigt und keine anderen als die angegebenen Hilfsmittel und Quellen verwendet habe. Diese Arbeit wurde weder in gleicher noch in ähnlicher Form in einem anderen Prüfungsverfahren vorgelegt. Ich habe früher, außer den mit dem Zulassungsgesuch urkundlichen Graden, keine weiteren akademischen Grade erworben und zu erwerben gesucht. Würzburg, Lars Hofmann Content SUMMARY ................................................................................................................ IV ZUSAMMENFASSUNG ............................................................................................. V 1. INTRODUCTION ................................................................................................. 1 1.1. Molecular basics of cancer .......................................................................................... 1 1.2. Early research on tumorigenesis ................................................................................. 3 1.3. Developing
    [Show full text]
  • RAP1-Mediated MEK/ERK Pathway Defects in Kabuki Syndrome
    The Journal of Clinical Investigation RESEARCH ARTICLE RAP1-mediated MEK/ERK pathway defects in Kabuki syndrome Nina Bögershausen,1,2,3 I-Chun Tsai,4 Esther Pohl,1,2,3 Pelin Özlem Simsek Kiper,5 Filippo Beleggia,1,2,3 E. Ferda Percin,6 Katharina Keupp,1,2,3 Angela Matchan,7 Esther Milz,1,2,3 Yasemin Alanay,5,8 Hülya Kayserili,9 Yicheng Liu,1,2,3 Siddharth Banka,10 Andrea Kranz,11 Martin Zenker,12 Dagmar Wieczorek,13 Nursel Elcioglu,14 Paolo Prontera,15 Stanislas Lyonnet,16 Thomas Meitinger,17 A. Francis Stewart,11 Dian Donnai,10 Tim M. Strom,17,18 Koray Boduroglu,5 Gökhan Yigit,1,2,3 Yun Li,1,2,3 Nicholas Katsanis,4 and Bernd Wollnik1,2,3 1Institute of Human Genetics, 2Center for Molecular Medicine Cologne (CMMC), and 3Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany. 4Center for Human Disease Modeling and Department of Cell Biology, Duke University, Durham, North Carolina, USA. 5Pediatric Genetics Unit, Department of Pediatrics, Hacettepe University Medical Faculty, Ankara, Turkey. 6Department of Medical Genetics, Gazi University Faculty of Medicine, Ankara, Turkey. 7Oxford Gene Technology (OGT), Begbroke Science Park, Begbroke, Oxfordshire, United Kingdom. 8Pediatric Genetics, Department of Pediatrics, Acibadem University, School of Medicine, Istanbul, Turkey. 9Medical Genetics Department, Koç University, School of Medicine, Istanbul, Turkey. 10Department of Genetic Medicine, St. Mary’s Hospital, Manchester Academic Health Sciences Centre (MAHSC), University of Manchester, Manchester, United Kingdom. 11Genomics, Bio-Innovationszentrum, Dresden University of Technology, Dresden, Germany. 12Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany.
    [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]
  • The Adherens Junction Protein Afadin Is an AKT Substrate That Regulates Breast Cancer Cell Migration
    Published OnlineFirst November 22, 2013; DOI: 10.1158/1541-7786.MCR-13-0398 Molecular Cancer Signal Transduction Research The Adherens Junction Protein Afadin Is an AKT Substrate that Regulates Breast Cancer Cell Migration Sivan Elloul, Dmitriy Kedrin, Nicholas W. Knoblauch, Andrew H. Beck, and Alex Toker Abstract The PI3K–AKT signaling pathway regulates all phenotypes that contribute to progression of human cancers, including breast cancer. AKT mediates signal relay by phosphorylating numerous substrates, which are causally implicated in biologic responses such as cell growth, survival, metabolic reprogramming, migration, and invasion. Here a new AKT substrate is identified, the adherens junction protein Afadin, which is phosphorylated by AKT at Ser1718. Importantly, under conditions of physiologic IGF-1 signaling and oncogenic PI3K and AKT, Afadin is phosphorylated by all AKT isoforms, and this phosphorylation elicits a relocalization of Afadin from adherens junctions to the nucleus. Also, phosphorylation of Afadin increased breast cancer cell migration that was dependent on Ser1718 phosphorylation. Finally, nuclear localization of Afadin was observed in clinical breast cancer specimens, indicating that regulation of Afadin by the PI3K–AKT pathway has pathophysiologic significance. Implications: Phosphorylation of the adhesion protein Afadin by AKT downstream of the PI3K pathway, leads to redistribution of Afadin and controls cancer cell migration. Mol Cancer Res; 12(3); 464–76. Ó2013 AACR. Introduction of the proteins that function to transduce PI3K and Akt The phosphoinositide 3-kinase (PI3K) and Akt signaling signaling are enzymes with catalytic pockets, this pathway is pathway orchestrates virtually all aspects of epithelial and highly druggable and numerous phase I and II clinical trials tumor cell behavior, from initial transformation to dysplasia are underway with small-molecule inhibitors targeting PI3K and ultimately the dissemination of cancer cells to distant or Akt isoforms for single agent or combination therapy, metastatic sites (1).
    [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]
  • Genome-Wide Association Study of Brain Amyloid Deposition As Measured by Pittsburgh Compound-B (Pib)-PET Imaging
    Molecular Psychiatry (2021) 26:309–321 https://doi.org/10.1038/s41380-018-0246-7 ARTICLE Genome-wide association study of brain amyloid deposition as measured by Pittsburgh Compound-B (PiB)-PET imaging 1,2 3,4 5 1 3,4 1 Qi Yan ● Kwangsik Nho ● Jorge L. Del-Aguila ● Xingbin Wang ● Shannon L. Risacher ● Kang-Hsien Fan ● 6,7 8 7,9 6,7,8 1 Beth E. Snitz ● Howard J. Aizenstein ● Chester A. Mathis ● Oscar L. Lopez ● F. Yesim Demirci ● 1 6,7,8 3,4 Eleanor Feingold ● William E. Klunk ● Andrew J. Saykin ● for the Alzheimer’s Disease Neuroimaging 5 1,7,8 Initiative (ADNI) ● Carlos Cruchaga ● M. Ilyas Kamboh Received: 1 December 2017 / Accepted: 31 July 2018 / Published online: 25 October 2018 © The Author(s) 2018. This article is published with open access Abstract Deposition of amyloid plaques in the brain is one of the two main pathological hallmarks of Alzheimer’s disease (AD). Amyloid positron emission tomography (PET) is a neuroimaging tool that selectively detects in vivo amyloid deposition in the brain and is a reliable endophenotype for AD that complements cerebrospinal fluid biomarkers with regional information. We measured in vivo amyloid deposition in the brains of ~1000 subjects from three collaborative AD centers and ADNI 11 1234567890();,: 1234567890();,: using C-labeled Pittsburgh Compound-B (PiB)-PET imaging followed by meta-analysis of genome-wide association studies, first to our knowledge for PiB-PET, to identify novel genetic loci for this endophenotype. The APOE region showed the most significant association where several SNPs surpassed the genome-wide significant threshold, with APOE*4 being most significant (P-meta = 9.09E-30; β = 0.18).
    [Show full text]