GABP Transcription Factor Is Required for Development of Chronic Myelogenous Leukemia Via Its Control of PRKD2

Total Page:16

File Type:pdf, Size:1020Kb

GABP Transcription Factor Is Required for Development of Chronic Myelogenous Leukemia Via Its Control of PRKD2 GABP transcription factor is required for development of chronic myelogenous leukemia via its control of PRKD2 Zhong-Fa Yanga,b, Haojian Zhanga,c, Leyuan Mad, Cong Penga,e,f, Yaoyu Chena,g, Junling Wanga, Michael R. Greend, Shaoguang Lia, and Alan G. Rosmarina,1 aDivision of Hematology–Oncology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655; bSchool of Basic Medical Sciences, TaiShan Medical University, TaiAn 271016, China; cDivision of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana–Farber Cancer Institute, Boston, MA 02215; dHoward Hughes Medical Institute and Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655; eDivision of Hematology/Oncology, Children’s Hospital Boston, Boston, MA 02115; fDepartment of Pediatric Oncology, Dana–Farber Cancer Institute, Boston, MA 02115; and gOncology, Novartis Institutes for BioMedical Research; Cambridge, MA 02139 Edited by Dennis A. Carson, University of California at San Diego, La Jolla, CA, and approved December 21, 2012 (received for review August 3, 2012) Hematopoietic stem cells (HSCs) are the source of all blood lineages, of GABPα binds to aminoterminal ankyrin-like repeats of GABPβ, and HSCs must balance quiescence, self-renewal, and differentiation whereas the transcription activation domain is encoded in the to meet lifelong needs for blood cell development. Transformation GABPβ carboxyl terminus. GABP controls expression of genes of HSCs by the breakpoint cluster region-ABL tyrosine kinase (BCR- that are required for innate and acquired immunity, including ABL) oncogene causes chronic myelogenous leukemia (CML). The CD18; Elastase, Neutrophil Expressed (ELANE); and α4 integrin E-twenty six (ets) transcription factor GA binding protein (GABP) is in myeloid cells (4), and interleukin-7 receptor (IL7R) and Pax5 in a tetrameric transcription factor complex that contains GABPα and lymphocytes (5, 6). GABP also controls expression of genes that GABPβ proteins. Deletion in bone marrow of Gabpa, the gene that are required for cell cycle control, and for ribosomal and mito- encodes the DNA-binding component, caused cell cycle arrest in HSCs chondrial biogenesis (4). GABPA and profound loss of hematopoietic progenitor cells. Loss of Gabpα is a unique gene in the human and mouse genomes, β prevented development of CML, although mice continued to gener- and its product is the only protein that can recruit GABP to Gabpa MEDICAL SCIENCES ate BCR-ABL–expressing Gabpα-null cells for months that were seri- DNA. Deletion of mouse inactivates the Gabp complex, ally transplantable and contributed to all lineages in secondary and was shown to cause embryonic lethality (5, 7, 8). Conditional Gabpa fi recipients. A bioinformatic screen identified the serine-threonine ki- deletion of in mouse embryonic broblasts caused pro- α nase protein kinase D2 (PRKD2) as a potential effector of GABP in found G1S cell cycle arrest (8). Loss of Gabp in bone marrow HSCs. Prkd2 expression was markedly reduced in Gabpα-null HSCs caused myelodysplasia and profound loss of bone marrow pro- genitor cells, but reports differ regarding the specific effects of and progenitor cells. Reduced expression of PRKD2 or pharmacologic Gabpa inhibition decreased cell cycling, and PRKD2 rescued growth of Gabpα- deletion on HSCs (9, 10). – We show that disruption of Gabpa markedly reduced HSC cell null BCR-ABL expressing cells. Thus, GABP is required for HSC cell cycle α entry and CML development through its control of PRKD2. This offers cycle activity, and that Gabp loss prevented development of CML in BCR-ABL–expressing bone marrow. Rather than developing a potential therapeutic target in leukemia. + leukemia, Gabpα-null BCR-ABL HSCs continued to generate + mature granulocytes for many months. Gabpα-null BCR-ABL LSC | cell cycle control | signal transduction | imatinib HSCs were transplantable into secondary recipients and contributed to all hematopoietic lineages. A bioinformatic screen implicated the ematopoietic stem cells (HSCs) are the source of all blood diacylglycerol- and protein kinase C (PKC)-activated serine-thre- Hlineages in bone marrow and peripheral blood. HSCs must onine kinase protein kinase D2 (PRKD2) as a potential effector of balance quiescence, growth, and differentiation to meet lifelong GABP in CML. Knockdown or pharmacologic inhibition of demands for blood cell development. HSCs give rise to lineage- PRKD2 mimicked the effect of Gabpa disruption on the growth of committed progenitor cells, yet retain the ability to renew the Gabpα-null HSCs and, conversely, ectopic expression of PRKD2 HSC pool. Transcription factors affect HSC proliferation, sur- overcame the growth defect of BCR-ABL–expressing Gabpα-null vival, and differentiation, and are implicated in leukemic trans- HSCs. Thus, Gabpα loss and expression of BCR-ABL achieve formation of HSCs (1). Chronic myelogenous leukemia (CML) is a myeloproliferative a standoff of sorts, i.e., the proliferative thrust of BCR-ABL par- fi tially overcomes the cell cycle arrest of Gabpα loss, whereas Gabpa neoplasm (MPN) characterized by in ltration of bone marrow, – peripheral blood, and viscera by myeloid cells. In CML, reciprocal disruption prevents BCR-ABL associated CML. This report translocation of chromosomes 9 and 22 generates the breakpoint describes a cell cycle control mechanism that prevents de- cluster region-ABL tyrosine kinase (BCR-ABL) oncogene. CML velopment of leukemia despite continued production of oncogene- arises in a leukemic stem cell (LSC), which drives the expansion expressing stem cells, and reports PRKD2 as a mediator of BCR- of granulocytes and their precursors that is the hallmark of this ABL transformation in CML. These findings identify a therapeutic MPN. BCR-ABL is a constitutively active tyrosine kinase that target in CML and strategies to prevent development of leukemia activates downstream signal transduction pathways, and inhibition in oncogene-expressing hematopoietic cells. of BCR-ABL by tyrosine kinase inhibitors (TKIs), such as ima- tinib, induces hematologic, cytogenetic, and molecular remission in most patients with CML (2). A mouse model of CML reca- Author contributions: Z.-F.Y., H.Z., L.M., C.P., Y.C., M.R.G., S.L., and A.G.R. designed re- pitulates aspects of human CML, and mice die within weeks of search; Z.-F.Y., H.Z., L.M., C.P., Y.C., and J.W. performed research; Z.-F.Y., H.Z., L.M., C.P., transplantation with BCR-ABL–expressing bone marrow (3). Y.C., M.R.G., and S.L. contributed new reagents/analytic tools; Z.-F.Y., H.Z., L.M., M.R.G., GA binding protein (GABP) is a tetrameric transcription factor S.L., and A.G.R. analyzed data; and Z.-F.Y. and A.G.R. wrote the paper. that contains two molecules of GABPα and two molecules of The authors declare no conflict of interest. various GABPβ proteins. GABP is unique among more than two This article is a PNAS Direct Submission. dozen mammalian E-twenty six (ets) transcription factors because 1To whom correspondence should be addressed. E-mail: [email protected]. it is the only obligate multimer, i.e., it is active only when the tet- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. ramer is formed. The carboxyl-terminal ets DNA binding domain 1073/pnas.1212904110/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1212904110 PNAS Early Edition | 1of6 Downloaded by guest on September 27, 2021 Results exhibited a profound loss of progenitor cells and a relative in- Gabpa Gabpa Deletion in Bone Marrow Causes Cell Cycle Arrest in HSCs. We crease in the percentage of HSCs. Disruption of caused fi P < created mice in which loxP recombination sites flank exons that a signi cant reduction in cell cycle activity in HSCs ( 0.01; Fig. fl/fl D fi encode the DNA-binding ets domain (Gabpa or floxed mice) S2 ). Thus, we con rmed the profound loss of bone marrow (8). The Mx1Cre transgene was bred into these mice, and injection progenitor cells, but preservation of primarily quiescent HSCs in with polyinosine-cytosine (pIC) deletes Gabpa in bone marrow Gabpα-null bone marrow. fl fl (Gabpa KO or simply KO mice) (9). As controls, Gabpa / lit- termates that lacked the Mx1Cre transgene were injected with Gabp Is Essential for Development of CML. In CML, transformation pIC. Half of the Gabpa KO mice died within 2 wk after the first of hematopoietic cells by BCR-ABL increases cellular prolifer- pIC injection (Fig. 1A). White blood cells, platelets, and hemo- ation and causes massive expansion of the granulocyte pool. globin in peripheral blood declined dramatically in KO mice be- Transplantation of mice with bone marrow cells that express BCR- tween days 5 and 14 (Fig. 1B), and microscopic examination ABL recapitulates many aspects of CML (3). We previously de- revealed bone marrow hemorrhage with only rare nucleated cells fined LSCs as BCR-ABL–expressing HSCs (11). Because loss of (Fig. 1C). However, peripheral blood counts in surviving KO mice Gabpα reduced HSC cell cycle activity, we sought to determine the recovered (Fig. 1B), and these mice lived for months without effect of Gabpa deletion on development of BCR-ABL–trans- apparent hematologic defects or other abnormalities. Only formed bone marrow. To delete Gabpa in BCR-ABL–expressing Gabpα-replete cells could form in vitro colonies when bone cells, we used a tricistronic retrovirus that expresses BCR-ABL, marrow was sampled 3 wk after pIC injection (Fig. S1). Thus, Cre recombinase, and green fluorescent protein (GFP) (12) to fl/fl rapid overgrowth of bone marrow by the initially small population infect WT or Gabpa bone marrow before transplantation (Fig. of cells that failed to delete Gabpa indicates that Gabpα-replete 2A). Expression of GFP permits identification of cells that both bone marrow cells have a growth advantage over Gabpα-null cells. express BCR-ABL and have undergone Cre-mediated deletion of Bone marrow HSCs are characterized by the absence of lineage floxed Gabpa.
Recommended publications
  • 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]
  • GAK and PRKCD Are Positive Regulators of PRKN-Independent
    bioRxiv preprint doi: https://doi.org/10.1101/2020.11.05.369496; this version posted November 5, 2020. 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-NC-ND 4.0 International license. 1 GAK and PRKCD are positive regulators of PRKN-independent 2 mitophagy 3 Michael J. Munson1,2*, Benan J. Mathai1,2, Laura Trachsel1,2, Matthew Yoke Wui Ng1,2, Laura 4 Rodriguez de la Ballina1,2, Sebastian W. Schultz2,3, Yahyah Aman4, Alf H. Lystad1,2, Sakshi 5 Singh1,2, Sachin Singh 2,3, Jørgen Wesche2,3, Evandro F. Fang4, Anne Simonsen1,2* 6 1Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo 7 2Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316, Oslo, Norway. 8 3Department of Molecular Cell Biology, The Norwegian Radium Hospital Montebello, N-0379, Oslo, Norway 9 4Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway 10 11 Keywords: GAK, Cyclin G Associated Kinase, PRKCD, Protein Kinase C Delta, Mitophagy, DFP, 12 DMOG, PRKN 13 14 *Corresponding Authors: 15 [email protected] 16 [email protected] 17 bioRxiv preprint doi: https://doi.org/10.1101/2020.11.05.369496; this version posted November 5, 2020. 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]
  • Supporting Information
    Supporting Information Chu et al. 10.1073/pnas.1613884113 Antibodies (Ter-119), anti-Ckit (2B8), anti-Sca1 (D7), and anti-CD64 (clone The following anti-mouse antibodies were purchased from X54-5/7.1). Anti-CD95 (15A7), anti-CD14 (clone Sa2-8), and anti- Biolegend: anti-B220 (RA3-6B2), anti-CD19 (6D5), anti-CD138 Tnfrsf13b (ebio8F10-3) antibodies were ordered from eBioscience. (281-2), anti-CD22 (OX-97), anti-CD24 (M1/69), anti-CD44 Anti-Cas9 (7A9-3A3) antibody was purchased from Cell Signaling (IM7), anti-CD80 (16-10A1), anti-CD81 (Eat-2), anti-CD83 Technology, and anti-Flag (M2) antibody and anti–β-actin were (Michel-19), anti-CD86 (GL-1), anti-CD45 (30-F11), anti-CD11b purchased from Sigma. HRP-conjugated secondary anti-mouse Ig (M1/70), anti-Gr-1 (RB6-8C5), anti-CD3e (145-2C11), anti-Ter-119 antibody was ordered from Rockland Immunochemicals. A In the mLN B cells Myeloid cells T cells C57BL/6 R26-Cas9iGFP/+ CD3 CD19 CD11b Cas9-iGFP B CD19+B220+ splenic B cells B2 B cells B1 B cells C57BL/6 B1: 4.1 R26-Cas9iGFP/+ CD43 CD19 B2: 95 CD5 Cas9-iGFP CD19+ peritoneal B cells B220loCD43+ B1 B2 B cells B1a B cells B1b B cells B1: 80 B1a: 64 CD5 CD19 CD43 B2: B1b: 20 31 B220 IgM Cas9-iGFP C Spleen CD19+B220+ CD38loFashi GC 55.4 CD19 CD38 CD19 1.2 B220 Fas Cas9-iGFP mLN 40.1 C57BL/6 R26-Cas9iGFP/+ CD19 CD38 3.8 CD19 B220 Fas Cas9-iGFP + + + Fig. S1. Characterization of heterozygous R26-Cas9iGFP transgenic mice.
    [Show full text]
  • ©Ferrata Storti Foundation
    Original Articles T-cell/histiocyte-rich large B-cell lymphoma shows transcriptional features suggestive of a tolerogenic host immune response Peter Van Loo,1,2,3 Thomas Tousseyn,4 Vera Vanhentenrijk,4 Daan Dierickx,5 Agnieszka Malecka,6 Isabelle Vanden Bempt,4 Gregor Verhoef,5 Jan Delabie,6 Peter Marynen,1,2 Patrick Matthys,7 and Chris De Wolf-Peeters4 1Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium; 2Department of Human Genetics, K.U.Leuven, Leuven, Belgium; 3Bioinformatics Group, Department of Electrical Engineering, K.U.Leuven, Leuven, Belgium; 4Department of Pathology, University Hospitals K.U.Leuven, Leuven, Belgium; 5Department of Hematology, University Hospitals K.U.Leuven, Leuven, Belgium; 6Department of Pathology, The Norwegian Radium Hospital, University of Oslo, Oslo, Norway, and 7Department of Microbiology and Immunology, Rega Institute for Medical Research, K.U.Leuven, Leuven, Belgium Citation: Van Loo P, Tousseyn T, Vanhentenrijk V, Dierickx D, Malecka A, Vanden Bempt I, Verhoef G, Delabie J, Marynen P, Matthys P, and De Wolf-Peeters C. T-cell/histiocyte-rich large B-cell lymphoma shows transcriptional features suggestive of a tolero- genic host immune response. Haematologica. 2010;95:440-448. doi:10.3324/haematol.2009.009647 The Online Supplementary Tables S1-5 are in separate PDF files Supplementary Design and Methods One microgram of total RNA was reverse transcribed using random primers and SuperScript II (Invitrogen, Merelbeke, Validation of microarray results by real-time quantitative Belgium), as recommended by the manufacturer. Relative reverse transcriptase polymerase chain reaction quantification was subsequently performed using the compar- Ten genes measured by microarray gene expression profil- ative CT method (see User Bulletin #2: Relative Quantitation ing were validated by real-time quantitative reverse transcrip- of Gene Expression, Applied Biosystems).
    [Show full text]
  • NRF1) Coordinates Changes in the Transcriptional and Chromatin Landscape Affecting Development and Progression of Invasive Breast Cancer
    Florida International University FIU Digital Commons FIU Electronic Theses and Dissertations University Graduate School 11-7-2018 Decipher Mechanisms by which Nuclear Respiratory Factor One (NRF1) Coordinates Changes in the Transcriptional and Chromatin Landscape Affecting Development and Progression of Invasive Breast Cancer Jairo Ramos [email protected] Follow this and additional works at: https://digitalcommons.fiu.edu/etd Part of the Clinical Epidemiology Commons Recommended Citation Ramos, Jairo, "Decipher Mechanisms by which Nuclear Respiratory Factor One (NRF1) Coordinates Changes in the Transcriptional and Chromatin Landscape Affecting Development and Progression of Invasive Breast Cancer" (2018). FIU Electronic Theses and Dissertations. 3872. https://digitalcommons.fiu.edu/etd/3872 This work is brought to you for free and open access by the University Graduate School at FIU Digital Commons. It has been accepted for inclusion in FIU Electronic Theses and Dissertations by an authorized administrator of FIU Digital Commons. For more information, please contact [email protected]. FLORIDA INTERNATIONAL UNIVERSITY Miami, Florida DECIPHER MECHANISMS BY WHICH NUCLEAR RESPIRATORY FACTOR ONE (NRF1) COORDINATES CHANGES IN THE TRANSCRIPTIONAL AND CHROMATIN LANDSCAPE AFFECTING DEVELOPMENT AND PROGRESSION OF INVASIVE BREAST CANCER A dissertation submitted in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY in PUBLIC HEALTH by Jairo Ramos 2018 To: Dean Tomás R. Guilarte Robert Stempel College of Public Health and Social Work This dissertation, Written by Jairo Ramos, and entitled Decipher Mechanisms by Which Nuclear Respiratory Factor One (NRF1) Coordinates Changes in the Transcriptional and Chromatin Landscape Affecting Development and Progression of Invasive Breast Cancer, having been approved in respect to style and intellectual content, is referred to you for judgment.
    [Show full text]
  • PRODUCTS and SERVICES Target List
    PRODUCTS AND SERVICES Target list Kinase Products P.1-11 Kinase Products Biochemical Assays P.12 "QuickScout Screening Assist™ Kits" Kinase Protein Assay Kits P.13 "QuickScout Custom Profiling & Panel Profiling Series" Targets P.14 "QuickScout Custom Profiling Series" Preincubation Targets Cell-Based Assays P.15 NanoBRET™ TE Intracellular Kinase Cell-Based Assay Service Targets P.16 Tyrosine Kinase Ba/F3 Cell-Based Assay Service Targets P.17 Kinase HEK293 Cell-Based Assay Service ~ClariCELL™ ~ Targets P.18 Detection of Protein-Protein Interactions ~ProbeX™~ Stable Cell Lines Crystallization Services P.19 FastLane™ Structures ~Premium~ P.20-21 FastLane™ Structures ~Standard~ Kinase Products For details of products, please see "PRODUCTS AND SERVICES" on page 1~3. Tyrosine Kinases Note: Please contact us for availability or further information. Information may be changed without notice. Expression Protein Kinase Tag Carna Product Name Catalog No. Construct Sequence Accession Number Tag Location System HIS ABL(ABL1) 08-001 Full-length 2-1130 NP_005148.2 N-terminal His Insect (sf21) ABL(ABL1) BTN BTN-ABL(ABL1) 08-401-20N Full-length 2-1130 NP_005148.2 N-terminal DYKDDDDK Insect (sf21) ABL(ABL1) [E255K] HIS ABL(ABL1)[E255K] 08-094 Full-length 2-1130 NP_005148.2 N-terminal His Insect (sf21) HIS ABL(ABL1)[T315I] 08-093 Full-length 2-1130 NP_005148.2 N-terminal His Insect (sf21) ABL(ABL1) [T315I] BTN BTN-ABL(ABL1)[T315I] 08-493-20N Full-length 2-1130 NP_005148.2 N-terminal DYKDDDDK Insect (sf21) ACK(TNK2) GST ACK(TNK2) 08-196 Catalytic domain
    [Show full text]
  • Clinical, Molecular, and Immune Analysis of Dabrafenib-Trametinib
    Supplementary Online Content Chen G, McQuade JL, Panka DJ, et al. Clinical, molecular and immune analysis of dabrafenib-trametinib combination treatment for metastatic melanoma that progressed during BRAF inhibitor monotherapy: a phase 2 clinical trial. JAMA Oncology. Published online April 28, 2016. doi:10.1001/jamaoncol.2016.0509. eMethods. eReferences. eTable 1. Clinical efficacy eTable 2. Adverse events eTable 3. Correlation of baseline patient characteristics with treatment outcomes eTable 4. Patient responses and baseline IHC results eFigure 1. Kaplan-Meier analysis of overall survival eFigure 2. Correlation between IHC and RNAseq results eFigure 3. pPRAS40 expression and PFS eFigure 4. Baseline and treatment-induced changes in immune infiltrates eFigure 5. PD-L1 expression eTable 5. Nonsynonymous mutations detected by WES in baseline tumors This supplementary material has been provided by the authors to give readers additional information about their work. © 2016 American Medical Association. All rights reserved. Downloaded From: https://jamanetwork.com/ on 09/30/2021 eMethods Whole exome sequencing Whole exome capture libraries for both tumor and normal samples were constructed using 100ng genomic DNA input and following the protocol as described by Fisher et al.,3 with the following adapter modification: Illumina paired end adapters were replaced with palindromic forked adapters with unique 8 base index sequences embedded within the adapter. In-solution hybrid selection was performed using the Illumina Rapid Capture Exome enrichment kit with 38Mb target territory (29Mb baited). The targeted region includes 98.3% of the intervals in the Refseq exome database. Dual-indexed libraries were pooled into groups of up to 96 samples prior to hybridization.
    [Show full text]
  • Supporting Information
    Supporting Information Faltermeier et al. 10.1073/pnas.1521674112 A. FU-R1-R2-V5-SV40-BlasƟ-CGW UbiquiƟn-C CMV 5’ LTR R1 Kinase R2 V5 SV40 BlastiR 3’ LTR promoter promoter GFP B. Kinase (kDa) 210 CMV GFP WPR 105 promot E er 78 49 38 28 V5 293t cells Fig. S1. Lentivirus-mediated overexpression of V5-tagged kinases. (A) Full-length kinases were cloned into the FU-R1-R2-V5-SV40-Blasti-CGW lentiviral vector shown. R1 and R2 represent recombination sites required for recombination-based Gateway cloning. LTR, Long-terminal repeat. (B) Western blot showing expression of selected kinases in 293t cells detected by a V5 antibody. The molecular mass of each kinase is indicated in parentheses. A. vector SrcY529F 3 Cap8 cells 2 weeks 2 x107 Tail vein Y529F Src injecƟon SCID 1 Luciferase reporter BLI Radiance (p/sec/cm3/sr) B. Lungs from mice injected with Cap8-SrcY529F cells Mouse 1 Mouse 2 Fig. S2. SrcY529F promotes lung colonization when overexpressed in murine prostate cells. (A) Experimental design to demonstrate that expression of mu- tationally activated kinase SrcY529F in Cap8 cells promotes lung colonization. (B) Bright-field images of lungs removed from mice 3 wk after being injected with Cap8-SrcY529F cells. (Scale bars, 5 mm.) Faltermeier et al. www.pnas.org/cgi/content/short/1521674112 1of5 A. Lungs-NTRK2 210 105 NTRK2 78 49 38 V5 293t cells Metastases Ɵssue Lungs-EGFR 210 EGFR 105 78 49 38 V5 293t cells Metastases Ɵssues Lungs-Her2 210 105 HER2 78 49 38 V5 293t cells Metastases Ɵssues B.
    [Show full text]
  • Gene Symbol Accession Alias/Prev Symbol Official Full Name AAK1 NM 014911.2 KIAA1048, Dkfzp686k16132 AP2 Associated Kinase 1
    Gene Symbol Accession Alias/Prev Symbol Official Full Name AAK1 NM_014911.2 KIAA1048, DKFZp686K16132 AP2 associated kinase 1 (AAK1) AATK NM_001080395.2 AATYK, AATYK1, KIAA0641, LMR1, LMTK1, p35BP apoptosis-associated tyrosine kinase (AATK) ABL1 NM_007313.2 ABL, JTK7, c-ABL, p150 v-abl Abelson murine leukemia viral oncogene homolog 1 (ABL1) ABL2 NM_007314.3 ABLL, ARG v-abl Abelson murine leukemia viral oncogene homolog 2 (arg, Abelson-related gene) (ABL2) ACVR1 NM_001105.2 ACVRLK2, SKR1, ALK2, ACVR1A activin A receptor ACVR1B NM_004302.3 ACVRLK4, ALK4, SKR2, ActRIB activin A receptor, type IB (ACVR1B) ACVR1C NM_145259.2 ACVRLK7, ALK7 activin A receptor, type IC (ACVR1C) ACVR2A NM_001616.3 ACVR2, ACTRII activin A receptor ACVR2B NM_001106.2 ActR-IIB activin A receptor ACVRL1 NM_000020.1 ACVRLK1, ORW2, HHT2, ALK1, HHT activin A receptor type II-like 1 (ACVRL1) ADCK1 NM_020421.2 FLJ39600 aarF domain containing kinase 1 (ADCK1) ADCK2 NM_052853.3 MGC20727 aarF domain containing kinase 2 (ADCK2) ADCK3 NM_020247.3 CABC1, COQ8, SCAR9 chaperone, ABC1 activity of bc1 complex like (S. pombe) (CABC1) ADCK4 NM_024876.3 aarF domain containing kinase 4 (ADCK4) ADCK5 NM_174922.3 FLJ35454 aarF domain containing kinase 5 (ADCK5) ADRBK1 NM_001619.2 GRK2, BARK1 adrenergic, beta, receptor kinase 1 (ADRBK1) ADRBK2 NM_005160.2 GRK3, BARK2 adrenergic, beta, receptor kinase 2 (ADRBK2) AKT1 NM_001014431.1 RAC, PKB, PRKBA, AKT v-akt murine thymoma viral oncogene homolog 1 (AKT1) AKT2 NM_001626.2 v-akt murine thymoma viral oncogene homolog 2 (AKT2) AKT3 NM_181690.1
    [Show full text]
  • Molecular Spectrum of BRAF, NRAS and KRAS Gene Mutations in Plasma Cell Dyscrasias: Implication for MEK-ERK Pathway Activation
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 6, No. 27 Molecular spectrum of BRAF, NRAS and KRAS gene mutations in plasma cell dyscrasias: implication for MEK-ERK pathway activation Marta Lionetti1, Marzia Barbieri2, Katia Todoerti3, Luca Agnelli1, Simona Marzorati1, Sonia Fabris2, Gabriella Ciceri1, Serena Galletti1, Giulia Milesi2, Martina Manzoni1, Mara Mazzoni4, Angela Greco4, Giovanni Tonon5, Pellegrino Musto3, Luca Baldini1,2 and Antonino Neri1,2 1 Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy 2 Hematology Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy 3 Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Potenza, Italy 4 Molecular Mechanism Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy 5 Functional Genomics of Cancer Unit, Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy Correspondence to: Antonino Neri, email: [email protected] Correspondence to: Marta Lionetti, email: [email protected] Keywords: multiple myeloma, plasma cell leukemia, RAS, BRAF, next generation sequencing Received: April 17, 2015 Accepted: May 31, 2015 Published: June 10, 2015 This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author
    [Show full text]
  • Downloaded from Phosphositeplus
    bioRxiv preprint doi: https://doi.org/10.1101/2020.07.22.215897; this version posted July 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. July 21, 2020 Phosphoproteomic Identification of Vasopressin-Regulated Protein Kinases in Collecting Duct Cells Arnab Datta1,2, Chin-Rang Yang1, Karim Salhadar1, Chung-Lin Chou1, Viswanathan Raghuram1, and Mark A. Knepper1 1Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 2Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore 575018, Karnataka, India Running title: Vasopressin-regulated Kinases in Renal Collecting Duct Correspondence to Mark Knepper, MD, PhD, Division of Intramural Research, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892 Phone: 301-496-3064; Email: [email protected] Keywords: mpkCCD, GPCR signaling, V2 receptor signaling, Desmopressin bioRxiv preprint doi: https://doi.org/10.1101/2020.07.22.215897; this version posted July 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. ABSTRACT Background and Purpose: The peptide hormone vasopressin regulates water transport in the renal collecting duct largely via the V2 receptor, which triggers a cAMP-mediated activation of a protein kinase A (PKA)-dependent signaling network.
    [Show full text]
  • Discovery of Potent and Selective MRCK Inhibitors with Therapeutic
    Published OnlineFirst January 30, 2018; DOI: 10.1158/0008-5472.CAN-17-2870 Cancer Translational Science Research Discovery of Potent and Selective MRCK Inhibitors with Therapeutic Effect on Skin Cancer Mathieu Unbekandt1, Simone Belshaw2, Justin Bower2, Maeve Clarke2, Jacqueline Cordes2, Diane Crighton2, Daniel R. Croft2, Martin J. Drysdale2, Mathew J. Garnett3, Kathryn Gill2, Christopher Gray2, David A. Greenhalgh4, James A.M. Hall3, Jennifer Konczal2, Sergio Lilla5, Duncan McArthur2, Patricia McConnell2, Laura McDonald2, Lynn McGarry6, Heather McKinnon2, Carol McMenemy4, Mokdad Mezna2, Nicolas A. Morrice5, June Munro1, Gregory Naylor1, Nicola Rath1, Alexander W. Schuttelkopf€ 2, Mairi Sime2, and Michael F. Olson1,7 Abstract The myotonic dystrophy–related Cdc42-binding kinases an autophosphorylation site, enabling development of a phos- MRCKa and MRCKb contribute to the regulation of actin–myosin phorylation-sensitive antibody tool to report on MRCKa status in cytoskeleton organization and dynamics, acting in concert with tumor specimens. In a two-stage chemical carcinogenesis model the Rho-associated coiled-coil kinases ROCK1 and ROCK2. The of murine squamous cell carcinoma, topical treatments reduced absence of highly potent and selective MRCK inhibitors has MRCKa S1003 autophosphorylation and skin papilloma out- resulted in relatively little knowledge of the potential roles of growth. In parallel work, we validated a phospho-selective anti- these kinases in cancer. Here, we report the discovery of the body with the capability to monitor drug pharmacodynamics. azaindole compounds BDP8900 and BDP9066 as potent and Taken together, our findings establish an important oncogenic selective MRCK inhibitors that reduce substrate phosphorylation, role for MRCK in cancer, and they offer an initial preclinical proof leading to morphologic changes in cancer cells along with inhi- of concept for MRCK inhibition as a valid therapeutic strategy.
    [Show full text]