The Role of Protein Kinase N in Gastric Cancer

Total Page:16

File Type:pdf, Size:1020Kb

The Role of Protein Kinase N in Gastric Cancer The Role of Protein Kinase N in Gastric Cancer Lizbeth Minerva Jiménez Flores ADVERTIMENT. La consulta d’aquesta tesi queda condicionada a l’acceptació de les següents condicions d'ús: La difusió d’aquesta tesi per mitjà del servei TDX (www.tdx.cat) i a través del Dipòsit Digital de la UB (diposit.ub.edu) ha estat autoritzada pels titulars dels drets de propietat intel·lectual únicament per a usos privats emmarcats en activitats d’investigació i docència. No s’autoritza la seva reproducció amb finalitats de lucre ni la seva difusió i posada a disposició des d’un lloc aliè al servei TDX ni al Dipòsit Digital de la UB. No s’autoritza la presentació del seu contingut en una finestra o marc aliè a TDX o al Dipòsit Digital de la UB (framing). Aquesta reserva de drets afecta tant al resum de presentació de la tesi com als seus continguts. En la utilització o cita de parts de la tesi és obligat indicar el nom de la persona autora. ADVERTENCIA. La consulta de esta tesis queda condicionada a la aceptación de las siguientes condiciones de uso: La difusión de esta tesis por medio del servicio TDR (www.tdx.cat) y a través del Repositorio Digital de la UB (diposit.ub.edu) ha sido autorizada por los titulares de los derechos de propiedad intelectual únicamente para usos privados enmarcados en actividades de investigación y docencia. No se autoriza su reproducción con finalidades de lucro ni su difusión y puesta a disposición desde un sitio ajeno al servicio TDR o al Repositorio Digital de la UB. No se autoriza la presentación de su contenido en una ventana o marco ajeno a TDR o al Repositorio Digital de la UB (framing). Esta reserva de derechos afecta tanto al resumen de presentación de la tesis como a sus contenidos. En la utilización o cita de partes de la tesis es obligado indicar el nombre de la persona autora. WARNING. On having consulted this thesis you’re accepting the following use conditions: Spreading this thesis by the TDX (www.tdx.cat) service and by the UB Digital Repository (diposit.ub.edu) has been authorized by the titular of the intellectual property rights only for private uses placed in investigation and teaching activities. Reproduction with lucrative aims is not authorized nor its spreading and availability from a site foreign to the TDX service or to the UB Digital Repository. Introducing its content in a window or frame foreign to the TDX service or to the UB Digital Repository is not authorized (framing). Those rights affect to the presentation summary of the thesis as well as to its contents. In the using or citation of parts of the thesis it’s obliged to indicate the name of the author. The Role of Protein Kinase N in Gastric Cancer by Lizbeth Minerva Jiménez Flores A thesis submitted in fulfillment of the requirements for the title of PhD by the University of Barcelona PhD program in Biomedicine The thesis was carried out at Group of Biomedical Research in Digestive Tract Tumors Centre for Molecular Biology and Biochemistry Investigation in Nanomedice (CIBBIM-Nanomedicine) Vall d’Hebron Research Institute Director: Co-Director: Diego Arango del Corro José Higinio Dopeso González PhD candidate: Lizbeth Minerva Jiménez Flores Esta tesis doctoral ha sido apoyada por el gobierno de los Estados Unidos Mexicanos, a través del programa nacional de Becas en el extranjero (convocatoria de 2014 segundo periodo) Registro y Solicitud de Beca No. 269733/383724 Número de Becario 389474 2 A mis padres y a mi hermana 3 Abstract Gastric cancer (GC) is one of the most common types of cancer in the Western World and accounts for over 700,000 deaths every year worldwide. The prognosis is dismal, with an average 5-year survival rate of less than 20%, mainly because of late diagnosis, due to the early stages are clinically silent. The cause of GC is multifactorial, as infectious agents, enviromental or/and genetic factors. Based on Lauren’s histologic classification, there are 2 types of GC: intestinal (IGC) and diffuse (DGC). Diffuse carcinoma cells lacks cohesion and invade tissues independently or in small clusters, is more common in young patients and behaves more aggressively than the intestinal type. Recent findings published in Nature and Nature Genetics identified frequent hotspots (14-24%) mutations in the small GTPase RHOA in the diffuse type of gastric tumors. To investigate the mechanism that underlies downstream of RHOA we analyzed the capacity of the mutations more commonly found in gastric tumors to bind to different effectors. We observed that the mutations found in gastric tumors specifically affect the capacity of RHOA to bind to PKN effector family. Therefore, in this thesis we study and characterized the role of PKN1 in GC. We demonstrated that the downregulation of PKN1 with shRNA or the deletion mediated by CRISPR/Cas9 results in the increase of proliferation in the diffuse gastric cell lines “in vitro” and “in vivo”. Moreover, the opposite effect is observed when we overexpress the constitutively active form of PKN1 in diffuse gastric cell lines with moderate or low levels of PKN1. In addition, we use a novel mouse model with conditional expression in the gastric mucosa of the RHOA-Y42C mutation (the most frequent mutation found in DGC) to investigate the role of RHOA in gastric tumorogenesis initiated by either N-methyl-N-nitrosourea (MNU) or Apc mutations. The mice with expression of the RHOA-Y42C have a significant increase in the number of tumors indicating that RhoA-Y42C is important for progression of the gastric tumors. Finally, we performed a preclinical testing of a new therapeutic approach, such as dietary supplements of arachidonic acid, a well-established activator of PKN1. The work carried out in this thesis will shed light on the newly identified deregulation of RHOA-PKN signaling in gastric cancer and provide a solid rationale for therapeutic targeting of this pathway. 4 Resumen El cáncer gástrico (CG) es uno de los tipos de cáncer más comunes en el mundo occidental y representa más de 700,000 muertes cada año en todo el mundo. El pronóstico es bajo, con una tasa de supervivencia promedio de 5 años de menos del 20%, principalmente debido a un diagnóstico tardío, debido a que las etapas tempranas son clínicamente silenciosas. La causa del CG es multifactorial, como agentes infecciosos, factores ambientales y/o genéticos. Según la clasificación histológica de Lauren, hay 2 tipos de CG: intestinal (CGI) y difuso (CGD). Las células de carcinoma de tipo difuso carecen de cohesión e invaden los tejidos de forma independiente o en pequeños grupos, es más común en pacientes jóvenes y se comporta de forma más agresiva que el tipo intestinal. Hallazgos recientes publicados en Nature and Nature Genetics identificaron mutaciones frecuentes en sitios específicos o hotspots (14-24%) en la proteína GTPasa RHOA en los tumores gástricos de tipo difuso. Para investigar el mecanismo que subyace en la vía de señalización de RHOA, analizamos la capacidad de las mutaciones más recurrentes encontradas en tumores gástricos para unirse a sus diferentes efectores. Observamos que las mutaciones encontradas en los tumores gástricos afectan específicamente la capacidad de RHOA para unirse los efectores de la familia de proteínas kinasas PKN. Por lo tanto, en esta tesis estudiamos y caracterizamos el papel de PKN1 en el CG. Demostramos que la depleción de PKN1 mediante shRNA o la deleción mediada por CRISPR/Cas9 da como resultado el aumento de la proliferación en las líneas celulares gástricas difusas "in vitro" e "in vivo". Además, el efecto opuesto se observa cuando sobreexpresamos la forma constitutivamente activa de PKN1 en líneas celulares gástricas difusas con niveles de expresión moderados o bajos de PKN1. Así mismo, utilizamos un nuevo modelo murino con expresión condicional en la membrana mucosa gástrica de la mutación RHOA-Y42C (la mutación más frecuente encontrada en CGD) para investigar el papel de RHOA en la tumorogénesis gástrica iniciada por N-metil-N-nitrosourea (MNU) o por mutaciones de Apc. Los ratones con expresión de RHOA-Y42C presentaron un aumento significativo en el número de tumores, lo cual indica que RHOA-Y42C tiene un papel importante en la progresión de los tumores gástricos. Finalmente, realizamos una prueba preclínica de un nuevo enfoque terapéutico, como los suplementos dietéticos de ácido araquidónico, un activador bien establecido de PKN1. El trabajo llevado a cabo en esta tesis destacará la importancia de la desregulación recientemente identificada de la señalización RHOA-PKN en el cáncer gástrico y proporcionará una base sólida para la orientación terapéutica de esta vía. 5 INDEX Abstract ....................................................................................................................................... 4 INDEX ......................................................................................................................................... 6 Figure index ........................................................................................................................... 10 Table index ............................................................................................................................ 12 CHAPTER I. Introduction ........................................................................................................... 13 1. The human gastrointestinal system ....................................................................................... 14 1.1 Overview .........................................................................................................................
Recommended publications
  • Targeting the LKB1 Tumor Suppressor
    Send Orders for Reprints to [email protected] 32 Current Drug Targets, 2014, 15, 32-52 Targeting the LKB1 Tumor Suppressor Rui-Xun Zhao and Zhi-Xiang Xu* Division of Hematology and Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA Abstract: LKB1 (also known as serine-threonine kinase 11, STK11) is a tumor suppressor, which is mutated or deleted in Peutz-Jeghers syndrome (PJS) and in a variety of cancers. Physiologically, LKB1 possesses multiple cellular functions in the regulation of cell bioenergetics metabolism, cell cycle arrest, embryo development, cell polarity, and apoptosis. New studies demonstrated that LKB1 may also play a role in the maintenance of function and dynamics of hematopoietic stem cells. Over the past years, personalized therapy targeting specific genetic aberrations has attracted intense interests. Within this review, several agents with potential activity against aberrant LKB1 signaling have been discussed. Potential strate- gies and challenges in targeting LKB1 inactivation are also considered. Keywords: LKB1 (serine-threonine kinase 11, STK11), AMP-activated protein kinase (AMPK), tumor suppression, mutations, targeting therapeutics. INTRODUCTION THE BIOLOGICAL FUNCTIONS OF LKB1 The LKB1 gene, also known as serine-threonine kinase Cell Metabolism 11 (STK11), was first identified by Jun-ichi Nezu of Chugai About a decade ago, studies from three different groups Pharmaceuticals in 1996 in a screen aimed at identifying new established that LKB1 is the long-sought kinase that phos- kinases [1]. The human LKB1 gene has been mapped to phorylates AMPK [9-11]. AMPK is a heterotrimeric enzyme chromosome 19p13.3. The gene spans 23 kb and is com- complex consisting of a catalytic subunit and regulatory posed of nine coding exons and a noncoding exon [2].
    [Show full text]
  • Overexpression and Selective Anticancer Efficacy of ENO3 in STK11 Mutant Lung Cancers
    Molecules and Cells Overexpression and Selective Anticancer Efficacy of ENO3 in STK11 Mutant Lung Cancers Choa Park1,3, Yejin Lee1,3, Soyeon Je1,3, Shengzhi Chang1, Nayoung Kim1, Euna Jeong2, and Sukjoon 1,2, Yoon * 1Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea, 2Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea, 3These authors contributed equally to this work. *Correspondence: [email protected] https://doi.org/10.14348/molcells.2019.0099 www.molcells.org Oncogenic gain-of-function mutations are clinical biomarkers lung cancer, constituting ~80% to 90% of all lung can- for most targeted therapies, as well as represent direct targets cers (Novello et al., 2016; Planchard et al., 2018). NSCLC for drug treatment. Although loss-of-function mutations is classified into three types: lung adenocarcinoma (LUAD), involving the tumor suppressor gene, STK11 (LKB1) are squamous cell carcinoma, and large cell carcinoma. LUAD important in lung cancer progression, STK11 is not the direct accounts for approximately 30% of lung cancers (Gill et al., target for anticancer agents. We attempted to identify cancer 2011). Serine/threonine kinase 11 (STK11), also known as liv- transcriptome signatures associated with STK11 loss-of- er kinase B1 (LKB1), is a major tumor suppressor gene in lung function mutations. Several new sensitive and specific gene cancers. Particularly, STK11 is the most inactivated tumor expression markers (ENO3, TTC39C, LGALS3, and MAML2) suppressor gene in NSCLC (Carretero et al., 2004; Chen et were identified using two orthogonal measures, i.e., fold al., 2016; Facchinetti et al., 2017). Although STK11 inactiva- change and odds ratio analyses of transcriptome data from tion (i.e., loss-of-function mutation) occurs in ~30% of LUAD cell lines and tissue samples.
    [Show full text]
  • Transcriptomic Analysis of Native Versus Cultured Human and Mouse Dorsal Root Ganglia Focused on Pharmacological Targets Short
    bioRxiv preprint doi: https://doi.org/10.1101/766865; this version posted September 12, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. Transcriptomic analysis of native versus cultured human and mouse dorsal root ganglia focused on pharmacological targets Short title: Comparative transcriptomics of acutely dissected versus cultured DRGs Andi Wangzhou1, Lisa A. McIlvried2, Candler Paige1, Paulino Barragan-Iglesias1, Carolyn A. Guzman1, Gregory Dussor1, Pradipta R. Ray1,#, Robert W. Gereau IV2, # and Theodore J. Price1, # 1The University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson, TX, 75080, USA 2Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine # corresponding authors [email protected], [email protected] and [email protected] Funding: NIH grants T32DA007261 (LM); NS065926 and NS102161 (TJP); NS106953 and NS042595 (RWG). The authors declare no conflicts of interest Author Contributions Conceived of the Project: PRR, RWG IV and TJP Performed Experiments: AW, LAM, CP, PB-I Supervised Experiments: GD, RWG IV, TJP Analyzed Data: AW, LAM, CP, CAG, PRR Supervised Bioinformatics Analysis: PRR Drew Figures: AW, PRR Wrote and Edited Manuscript: AW, LAM, CP, GD, PRR, RWG IV, TJP All authors approved the final version of the manuscript. 1 bioRxiv preprint doi: https://doi.org/10.1101/766865; this version posted September 12, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • Application of a MYC Degradation
    SCIENCE SIGNALING | RESEARCH ARTICLE CANCER Copyright © 2019 The Authors, some rights reserved; Application of a MYC degradation screen identifies exclusive licensee American Association sensitivity to CDK9 inhibitors in KRAS-mutant for the Advancement of Science. No claim pancreatic cancer to original U.S. Devon R. Blake1, Angelina V. Vaseva2, Richard G. Hodge2, McKenzie P. Kline3, Thomas S. K. Gilbert1,4, Government Works Vikas Tyagi5, Daowei Huang5, Gabrielle C. Whiten5, Jacob E. Larson5, Xiaodong Wang2,5, Kenneth H. Pearce5, Laura E. Herring1,4, Lee M. Graves1,2,4, Stephen V. Frye2,5, Michael J. Emanuele1,2, Adrienne D. Cox1,2,6, Channing J. Der1,2* Stabilization of the MYC oncoprotein by KRAS signaling critically promotes the growth of pancreatic ductal adeno- carcinoma (PDAC). Thus, understanding how MYC protein stability is regulated may lead to effective therapies. Here, we used a previously developed, flow cytometry–based assay that screened a library of >800 protein kinase inhibitors and identified compounds that promoted either the stability or degradation of MYC in a KRAS-mutant PDAC cell line. We validated compounds that stabilized or destabilized MYC and then focused on one compound, Downloaded from UNC10112785, that induced the substantial loss of MYC protein in both two-dimensional (2D) and 3D cell cultures. We determined that this compound is a potent CDK9 inhibitor with a previously uncharacterized scaffold, caused MYC loss through both transcriptional and posttranslational mechanisms, and suppresses PDAC anchorage- dependent and anchorage-independent growth. We discovered that CDK9 enhanced MYC protein stability 62 through a previously unknown, KRAS-independent mechanism involving direct phosphorylation of MYC at Ser .
    [Show full text]
  • Genome-Wide Sirna Screen for Mediators of NF-Κb Activation
    Genome-wide siRNA screen for mediators SEE COMMENTARY of NF-κB activation Benjamin E. Gewurza, Fadi Towficb,c,1, Jessica C. Marb,d,1, Nicholas P. Shinnersa,1, Kaoru Takasakia, Bo Zhaoa, Ellen D. Cahir-McFarlanda, John Quackenbushe, Ramnik J. Xavierb,c, and Elliott Kieffa,2 aDepartment of Medicine and Microbiology and Molecular Genetics, Channing Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115; bCenter for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114; cProgram in Medical and Population Genetics, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142; dDepartment of Biostatistics, Harvard School of Public Health, Boston, MA 02115; and eDepartment of Biostatistics and Computational Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115 Contributed by Elliott Kieff, December 16, 2011 (sent for review October 2, 2011) Although canonical NFκB is frequently critical for cell proliferation, (RIPK1). TRADD engages TNFR-associated factor 2 (TRAF2), survival, or differentiation, NFκB hyperactivation can cause malig- which recruits the ubiquitin (Ub) E2 ligase UBC5 and the E3 nant, inflammatory, or autoimmune disorders. Despite intensive ligases cIAP1 and cIAP2. CIAP1/2 polyubiquitinate RIPK1 and study, mammalian NFκB pathway loss-of-function RNAi analyses TRAF2, which recruit and activate the K63-Ub binding proteins have been limited to specific protein classes. We therefore under- TAB1, TAB2, and TAB3, as well as their associated kinase took a human genome-wide siRNA screen for novel NFκB activa- MAP3K7 (TAK1). TAK1 in turn phosphorylates IKKβ activa- tion pathway components. Using an Epstein Barr virus latent tion loop serines to promote IKK activity (4).
    [Show full text]
  • STK11 Gene Serine/Threonine Kinase 11
    STK11 gene serine/threonine kinase 11 Normal Function The STK11 gene (also called LKB1) provides instructions for making an enzyme called serine/threonine kinase 11. This enzyme is a tumor suppressor, which means that it helps keep cells from growing and dividing too fast or in an uncontrolled way. This enzyme helps certain types of cells correctly orient themselves within tissues ( polarization) and assists in determining the amount of energy a cell uses. This kinase also promotes a type of programmed cell death known as apoptosis. In addition to its role as a tumor suppressor, serine/threonine kinase 11 function appears to be required for normal development before birth. Health Conditions Related to Genetic Changes Breast cancer Inherited changes in the STK11 gene greatly increase the risk of developing breast cancer, as well as other types of cancer, as part of Peutz-Jeghers syndrome (described above). These mutations are thought to account for only a small fraction of all breast cancer cases. Peutz-Jeghers syndrome Inherited mutations in the STK11 gene cause Peutz-Jeghers syndrome, a condition characterized by the development of noncancerous growths called hamartomatous polyps in the gastrointestinal tract and a greatly increased risk of developing several types of cancer. More than 340 STK11 gene mutations have been identified in people with this condition. Many of these mutations result in the production of an abnormally short, nonfunctional version of the serine/threonine kinase 11 enzyme. Other mutations change single protein building blocks (amino acids) used to build the enzyme. Mutations in the STK11 gene impair the enzyme's tumor suppressor function, allowing cells to grow and divide without control or order.
    [Show full text]
  • Supplementary Table 1
    SI Table S1. Broad protein kinase selectivity for PF-2771. Kinase, PF-2771 % Inhibition at 10 μM Service Kinase, PF-2771 % Inhibition at 1 μM Service rat RPS6KA1 (RSK1) 39 Dundee AURKA (AURA) 24 Invitrogen IKBKB (IKKb) 26 Dundee CDK2 /CyclinA 21 Invitrogen mouse LCK 25 Dundee rabbit MAP2K1 (MEK1) 19 Dundee AKT1 (AKT) 21 Dundee IKBKB (IKKb) 16 Dundee CAMK1 (CaMK1a) 19 Dundee PKN2 (PRK2) 14 Dundee RPS6KA5 (MSK1) 18 Dundee MAPKAPK5 14 Dundee PRKD1 (PKD1) 13 Dundee PIM3 12 Dundee MKNK2 (MNK2) 12 Dundee PRKD1 (PKD1) 12 Dundee MARK3 10 Dundee NTRK1 (TRKA) 12 Invitrogen SRPK1 9 Dundee MAPK12 (p38g) 11 Dundee MAPKAPK5 9 Dundee MAPK8 (JNK1a) 11 Dundee MAPK13 (p38d) 8 Dundee rat PRKAA2 (AMPKa2) 11 Dundee AURKB (AURB) 5 Dundee NEK2 11 Invitrogen CSK 5 Dundee CHEK2 (CHK2) 11 Invitrogen EEF2K (EEF-2 kinase) 4 Dundee MAPK9 (JNK2) 9 Dundee PRKCA (PKCa) 4 Dundee rat RPS6KA1 (RSK1) 8 Dundee rat PRKAA2 (AMPKa2) 4 Dundee DYRK2 7 Dundee rat CSNK1D (CKId) 3 Dundee AKT1 (AKT) 7 Dundee LYN 3 BioPrint PIM2 7 Invitrogen CSNK2A1 (CKIIa) 3 Dundee MAPK15 (ERK7) 6 Dundee CAMKK2 (CAMKKB) 1 Dundee mouse LCK 5 Dundee PIM3 1 Dundee PDPK1 (PDK1) (directed 5 Invitrogen rat DYRK1A (MNB) 1 Dundee RPS6KB1 (p70S6K) 5 Dundee PBK 0 Dundee CSNK2A1 (CKIIa) 4 Dundee PIM1 -1 Dundee CAMKK2 (CAMKKB) 4 Dundee DYRK2 -2 Dundee SRC 4 Invitrogen MAPK12 (p38g) -2 Dundee MYLK2 (MLCK_sk) 3 Invitrogen NEK6 -3 Dundee MKNK2 (MNK2) 2 Dundee RPS6KB1 (p70S6K) -3 Dundee SRPK1 2 Dundee AKT2 -3 Dundee MKNK1 (MNK1) 2 Dundee RPS6KA3 (RSK2) -3 Dundee CHEK1 (CHK1) 2 Invitrogen rabbit MAP2K1 (MEK1) -4 Dundee
    [Show full text]
  • Pancancer Progression Human Vjune2017
    Gene Symbol Accession Alias/Prev Symbol Official Full Name AAMP NM_001087.3 - angio-associated, migratory cell protein ABI3BP NM_015429.3 NESHBP|TARSH ABI family, member 3 (NESH) binding protein ACHE NM_000665.3 ACEE|ARACHE|N-ACHE|YT acetylcholinesterase ACTG2 NM_001615.3 ACT|ACTA3|ACTE|ACTL3|ACTSG actin, gamma 2, smooth muscle, enteric ACVR1 NM_001105.2 ACTRI|ACVR1A|ACVRLK2|ALK2|FOP|SKR1|TSRI activin A receptor, type I ACVR1C NM_145259.2 ACVRLK7|ALK7 activin A receptor, type IC ACVRL1 NM_000020.1 ACVRLK1|ALK-1|ALK1|HHT|HHT2|ORW2|SKR3|TSR-I activin A receptor type II-like 1 ADAM15 NM_207195.1 MDC15 ADAM metallopeptidase domain 15 ADAM17 NM_003183.4 ADAM18|CD156B|CSVP|NISBD|TACE ADAM metallopeptidase domain 17 ADAM28 NM_014265.4 ADAM 28|ADAM23|MDC-L|MDC-Lm|MDC-Ls|MDCL|eMDC II|eMDCII ADAM metallopeptidase domain 28 ADAM8 NM_001109.4 CD156|MS2 ADAM metallopeptidase domain 8 ADAM9 NM_001005845.1 CORD9|MCMP|MDC9|Mltng ADAM metallopeptidase domain 9 ADAMTS1 NM_006988.3 C3-C5|METH1 ADAM metallopeptidase with thrombospondin type 1 motif, 1 ADAMTS12 NM_030955.2 PRO4389 ADAM metallopeptidase with thrombospondin type 1 motif, 12 ADAMTS8 NM_007037.4 ADAM-TS8|METH2 ADAM metallopeptidase with thrombospondin type 1 motif, 8 ADAP1 NM_006869.2 CENTA1|GCS1L|p42IP4 ArfGAP with dual PH domains 1 ADD1 NM_001119.4 ADDA adducin 1 (alpha) ADM2 NM_001253845.1 AM2|dJ579N16.4 adrenomedullin 2 ADRA2B NM_000682.4 ADRA2L1|ADRA2RL1|ADRARL1|ALPHA2BAR|alpha-2BAR adrenoceptor alpha 2B AEBP1 NM_001129.3 ACLP AE binding protein 1 AGGF1 NM_018046.3 GPATC7|GPATCH7|HSU84971|HUS84971|VG5Q
    [Show full text]
  • Atu027, a Liposomal Small Interfering RNA Formulation Targeting Protein Kinase N3, Inhibits Cancer Progression
    Research Article Atu027, a Liposomal Small Interfering RNA Formulation Targeting Protein Kinase N3, Inhibits Cancer Progression Manuela Aleku,1 Petra Schulz,2 Oliver Keil,1 Ansgar Santel,1 Ute Schaeper,1 Britta Dieckhoff,1 Oliver Janke,1 Jens Endruschat,1 Birgit Durieux,1 Nadine Ro¨der,1 Kathrin Lo¨ffler,1 Christian Lange,1 Melanie Fechtner,1 Kristin Mo¨pert,1 Gerald Fisch,1 Sibylle Dames,1 Wolfgang Arnold,1 Karin Jochims,3 Klaus Giese,1 Bertram Wiedenmann,2 Arne Scholz,2 and Jo¨rg Kaufmann1 1Silence Therapeutics AG; 2Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Charite´-Universita¨tsmedizin, Berlin, Germany; and 3IASON Consulting, Niederzier, Germany Abstract of chemically synthesized siRNA for therapeutic purposes (for a We have previously described a small interfering RNA (siRNA) technology review, see refs. 2, 4, 6–8). With respect to cancer, a delivery system (AtuPLEX) for RNA interference (RNAi) in the variety of nonviral nanoparticles (50–200 nm) have been recently vasculature of mice. Here we report preclinical data for reported to be suitable for RNAi-mediated tumor growth inhibition Atu027, a siRNA-lipoplex directed against protein kinase N3 in experimental mouse models (summarized in ref. 6). In most (PKN3), currently under development for the treatment of cases, various siRNA carriers such as cyclodextrin (transferrin advanced solid cancer. In vitro studies revealed that Atu027- receptor targeted; ref. 9), PEI (RGD-targeted PEG-PEI; ref. 10), mediated inhibition of PKN3 function in primary endothelial DOTAP (self-assembled LPD-nanoparticle; nanosized immunolipo- cells impaired tube formation on extracellular matrix and some; refs. 11, 12), AtuFECT (AtuPLEX; refs.
    [Show full text]
  • UPSTREAM COMPONENTS of MTORC1 by Li Li a Dissertation
    UPSTREAM COMPONENTS OF MTORC1 by Li Li A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Biological Chemistry) in The University of Michigan 2011 Doctoral Committee: Professor Kunliang Guan, Chair Professor Randal J. Kaufman Associate Professor Anne B. Vojtek Associate Professor Martin G. Myers Assistant Professor Diane C. Fingar © Li Li 2011 This work is dedicated to my parents. Without their support and guidance, I would not be where I am today. I also dedicate this work to my husband, for pushing me to the best I can. Last, and most importantly, I would like to dedicate this work to my fifteen months son for being well behaved during the last few months. You are the treasure of my life. ii ACKNOWLEDGEMENTS I would like to express my deep gratitude to my mentor Dr. Kun-Liang Guan for his direction, encouragement, and support during my graduate study. I am very thankful to him for his patience with me, enduring my naïve questions. That Dr. Guan is so approachable and enthusiastic about science set me an example on how to be a great PI and a great scientist. Next, I sincerely acknowledge the members of my thesis committee, Drs Randal J. Kaufman, Anne B. Vojtek, Diane C. Fingar and Martin G. Myers for their insightful suggestions and constructive criticism on my work. I would like to thank the current and former members of the Guan lab. All of them made the lab a great place to work. Especially, I would like to thank Bin Zhao, my husband, also a Guan lab member, for teaching me every lab techniques.
    [Show full text]
  • Supplementary Table 1. in Vitro Side Effect Profiling Study for LDN/OSU-0212320. Neurotransmitter Related Steroids
    Supplementary Table 1. In vitro side effect profiling study for LDN/OSU-0212320. Percent Inhibition Receptor 10 µM Neurotransmitter Related Adenosine, Non-selective 7.29% Adrenergic, Alpha 1, Non-selective 24.98% Adrenergic, Alpha 2, Non-selective 27.18% Adrenergic, Beta, Non-selective -20.94% Dopamine Transporter 8.69% Dopamine, D1 (h) 8.48% Dopamine, D2s (h) 4.06% GABA A, Agonist Site -16.15% GABA A, BDZ, alpha 1 site 12.73% GABA-B 13.60% Glutamate, AMPA Site (Ionotropic) 12.06% Glutamate, Kainate Site (Ionotropic) -1.03% Glutamate, NMDA Agonist Site (Ionotropic) 0.12% Glutamate, NMDA, Glycine (Stry-insens Site) 9.84% (Ionotropic) Glycine, Strychnine-sensitive 0.99% Histamine, H1 -5.54% Histamine, H2 16.54% Histamine, H3 4.80% Melatonin, Non-selective -5.54% Muscarinic, M1 (hr) -1.88% Muscarinic, M2 (h) 0.82% Muscarinic, Non-selective, Central 29.04% Muscarinic, Non-selective, Peripheral 0.29% Nicotinic, Neuronal (-BnTx insensitive) 7.85% Norepinephrine Transporter 2.87% Opioid, Non-selective -0.09% Opioid, Orphanin, ORL1 (h) 11.55% Serotonin Transporter -3.02% Serotonin, Non-selective 26.33% Sigma, Non-Selective 10.19% Steroids Estrogen 11.16% 1 Percent Inhibition Receptor 10 µM Testosterone (cytosolic) (h) 12.50% Ion Channels Calcium Channel, Type L (Dihydropyridine Site) 43.18% Calcium Channel, Type N 4.15% Potassium Channel, ATP-Sensitive -4.05% Potassium Channel, Ca2+ Act., VI 17.80% Potassium Channel, I(Kr) (hERG) (h) -6.44% Sodium, Site 2 -0.39% Second Messengers Nitric Oxide, NOS (Neuronal-Binding) -17.09% Prostaglandins Leukotriene,
    [Show full text]
  • Analysis of 3-Phosphoinositide-Dependent Kinase-1 Signaling and Function in ES Cells
    EXPERIMENTAL CELL RESEARCH 314 (2008) 2299– 2312 available at www.sciencedirect.com www.elsevier.com/locate/yexcr Research Article Analysis of 3-phosphoinositide-dependent kinase-1 signaling and function in ES cells Tanja Tamgüneya,b, Chao Zhangc, Dorothea Fiedlerc, Kevan Shokatc, David Stokoea,⁎ aUCSF Cancer Research Institute, USA bMolecular Medicine Program, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany cDepartment of Cellular and Molecular Pharmacology, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94115, USA ARTICLE INFORMATION ABSTRACT Article Chronology: 3-Phosphoinositide-dependent kinase-1 (PDK1) phosphorylates and activates several Received 5 February 2008 kinases in the cAMP-dependent, cGMP-dependent and protein kinase C (AGC) family. Revised version received Many putative PDK1 substrates have been identified, but have not been analyzed following 15 April 2008 transient and specific inhibition of PDK1 activity. Here, we demonstrate that a previously Accepted 16 April 2008 characterized PDK1 inhibitor, BX-795, shows biological effects that are not consistent with Available online 23 April 2008 PDK1 inhibition. Therefore, we describe the creation and characterization of a PDK1 mutant, L159G, which can bind inhibitor analogues containing bulky groups that hinder access to the − − Keywords: ATP binding pocket of wild type (WT) kinases. When expressed in PDK1 / ES cells, PDK1 PDK1 L159G restored phosphorylation of PDK1 targets known to be hypophosphorylated in these PKB/Akt cells. Screening of multiple inhibitor analogues showed that 1-NM-PP1 and 3,4-DMB-PP1 − − PI3K optimally inhibited the phosphorylation of PDK1 targets in PDK1 / ES cells expressing PDK1 Chemical genetics L159G but not WT PDK1. These compounds confirmed previously assumed PDK1 substrates, AGC kinases but revealed distinct dephosphorylation kinetics.
    [Show full text]