Oncogenic Transformation Induced by Membrane-Targeted Akt2 and Akt3

Total Page:16

File Type:pdf, Size:1020Kb

Oncogenic Transformation Induced by Membrane-Targeted Akt2 and Akt3 Oncogene (2001) 20, 4419 ± 4423 ã 2001 Nature Publishing Group All rights reserved 0950 ± 9232/01 $15.00 www.nature.com/onc Oncogenic transformation induced by membrane-targeted Akt2 and Akt3 Ines Mende1,3, Scott Malstrom2, Philip N Tsichlis2, Peter K Vogt*,1 and Masahiro Aoki1 1Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California CA 92037, USA; 2Kimmel Cancer Center, Thomas Jeerson University, Philadelphia, Pennsylvania PA 19107, USA The kinases Akt2, Akt3 and their myristylated variants, v-P3k induces oncogenic transformation of chicken Myr-Akt2 and Myr-Akt3 were expressed by the RCAS embryo ®broblasts (CEF) in culture and hemangiosar- vector in chicken embryo ®broblasts (CEF). Myr-Akt2 comas in chickens (Chang et al., 1997). The transform- and Myr-Akt3 were strongly oncogenic, inducing multi- ing activity of the P3k proteins is routed through the layered foci of transformed cells. In contrast, wild-type serine-threonine kinase Akt1 (protein kinase Ba), Akt2 and Akt3 were only poorly transforming, their because dominant negative Akt1 interferes with P3k- eciencies of focus formation were more than 100-fold induced transformation (Aoki et al., 1998). The akt1 lower; foci appeared later and showed less multilayering. gene was originally isolated as the oncogene of the Addition of the myristylation signal not only enhanced murine lymphomagenic retrovirus AKT8 (Bellacosa et oncogenic potential but also increased kinase activities. al., 1991). Cellular Akt1 binds with its plekstrin Myr-Akt2 and Myr-Akt3 also induced hemangiosarco- homology (PH) domain to the product of PI3K, D3- mas in the animal, whereas wild type Akt2 and Akt3 PPI, and thus is translocated to the plasma membrane. were not oncogenic in vivo. Furthermore, Akt2, driven by At this location, the protein kinases PDK1 and PDK2 the lck (lymphocyte speci®c kinase) promoter in activate Akt1 by phosphorylation of threonine 308 and transgenic mice, induced lymphomas. The oncogenic serine 473 (Alessi and Cohen, 1998; Chan et al., 1999; eects of Akt2 and Akt3 described here are indis- Coer et al., 1998; Datta et al., 1999). Constitutively tinguishable from those of Akt1. The downstream targets activated and membrane-targeted Akt1 causes focus relevant to oncogenic transformation are therefore formation in CEF and hemangiosarcomas in chickens probably shared by the three Akt kinases. Oncogene identical to the tumors induced by P3k (Aoki et al., (2001) 20, 4419 ± 4423. 1998). The oncogenic activity of the PI3K-Akt path- way is also manifest in human cancer. P110a Keywords: Akt; transformation; serine/threonine kinase (PIK110CA) is ampli®ed and overexpressed in ovarian cancers (Shayesteh et al., 1999). Akt1 is ampli®ed in gastric cancer (Staal, 1987). The function of a negative Signals controlled by PI 3-kinase (PI3K) aect diverse regulator of PI3K and Akt signals, PTEN, is lost in cellular functions including response to growth factors, several types of human tumors including glioblastomas dierentiation, and cell survival (Alessi and Downes, and prostate cancer. This loss of PTEN function leads 1998; Fruman et al., 1998; Shepherd et al., 1998; to high levels of D3-PPI and activation of Akt1 (Ali et Wymann and Pirola, 1998). Oncoproteins of the al., 1999; Cantley and Neel, 1999; Di Cristofano and receptor tyrosine kinase and Src families as well as Pandol®, 2000; Maehama and Dixon, 1999). polyoma middle T antigen activate PI3K through In addition to akt1, there are two related genes in association with the regulatory subunit of PI3K, p85, mammalian genomes, akt2 and akt3. These genes code suggesting a role of PI3K in the actions of these for the Akt2 and Akt3 kinases which show a high proteins (Fukui and Hanafusa, 1989; Hu et al., 1992; degree of sequence homology in their catalytic and PH Peles et al., 1992; Reith et al., 1991; Whitman et al., domains to Akt1 but diverge from Akt1 in other 1985). The Ras oncoprotein can activate PI3K by domains. The two regulatory phosphorylation sites binding to the catalytic subunit directly (Rodriguez- that correspond to threonine 308 and serine 473 of ONCOGENOMICS Viciana et al., 1994, 1996). We previously showed that Akt1 are conserved, and Akt2 and Akt3 also transduce the oncoprotein v-P3k of the avian retroviruses ASV PI3K signals (Altomare et al., 1998; Brodbeck et al., 16 and ASV 8905 codes for the catalytic subunit of 1999; Liu et al., 1998; Masure et al., 1999; Meier et al., PI3K, p110a (Chang et al., 1997; Aoki et al., 2000). 1997; Nakatani et al., 1999a). For both Akt2 and Akt3 there are data suggesting a role in human cancer. Akt2 is ampli®ed and overexpressed in ovarian cancer, breast cancer and pancreatic cancer and Akt3 is *Correspondence: PK Vogt, 10550 N. Torrey Pines Rd., BCC-239, overexpressed in breast and prostate cancer (Bellacosa La Jolla, CA 92037, USA et al., 1995; Cheng et al., 1992, 1996; Miwa et al., 1996; 3 Current address: Universitatsklinikum Essen, Virchowstrasse 173, Nakatani et al., 1999b; Ruggeri et al., 1998). In this 45122 Essen, Germany Received 29 November 2000; revised 20 March 2001; accepted 26 communication, we describe oncogenic transformation March 2001 by Akt2 and Akt3 in experimental systems. The in vitro Neoplastic transformation by Akt2 and Akt3 I Mende et al 4420 transformation assays have obvious utility in screens transforming, about as inecient in focus formation for Akt antagonists. as non-myristylated Akt1 (Table 1). Foci induced by Akt2 (of murine origin) and Akt3 (from rat) were R-Akt2 or R-Akt3 took about 3 weeks to develop and cloned with an HA tag in the avian retroviral showed signi®cantly reduced multilayering. Rare in- expression vector RCAS to yield the constructs R- stances of highly transformed foci in R-Akt2 or R- Akt2 and R-Akt3 (Aoki et al., 1998; Hughes et al., Akt3-infected cultures probably resulted from muta- 1987). Two additional constructs expressed the same tions acquired during retroviral replication of the tagged Akt2 or Akt3 proteins but with the myristyla- constructs. As was shown in a previous study, such tion signal of the c-Src kinase added at the amino mutations occur during RCAS replication and can termini (R-Myr-Akt2 and R-Myr-Akt3). The pre- greatly enhance focus formation mediated by a cellular viously described RCAS constructs expressing Akt1 insert in RCAS (Aoki et al., 2000). Oncogenicity of and myristylated Akt1 (R-Akt1 and R-Myr-Akt1) non-myristylated Akt2 was observed previously by served as controls (Figure 1) (Aoki et al., 1998). another group using NIH3T3 cells (Cheng et al., 1997). Separate CEF cultures were transfected with these The apparent disagreement with our results may be constructs, and supernatant growth medium containing explained by the dierence in the host cells used in the infectious RCAS virus with the respective inserts was two studies. Western blots with anti HA epitope used in assays for focus formation on CEF. R-Myr- antibody detected high levels of all Akt proteins in Akt2 and R-Myr-Akt3 were strongly transforming, infected CEF (Figure 3a). The vestigial transforming inducing foci of multilayered cells within one week as activity of R-Akt2 or R-Akt3 was therefore not caused eciently as R-Myr-Akt1 (Figure 2, Table 1). In by insucient protein expression. Kinase activities contrast, the constructs expressing the wild-type were determined by immune complex kinase assays kinases, R-Akt2 and R-Akt3 were only weakly using anti HA antibody for immunoprecipitation and Figure 1 Schematic representation of the Akt constructs used in this study. The pleckstrin homology (PH) domain, protein kinase domain, the two regulatory phosphorylation sites, the myristylation signal, and the HA epitope tag are indicated. Constructs of Akt1 and the Myr-Akt1 mutant were described previously. Akt2, and Myr-Akt2 with the myristylation signal of c-Src proteins were subcloned into the avian retroviral vector RCAS.S® via the adapter vector pBSFI (Aoki et al., 1998). The incomplete carboxyl terminal sequences of the rat akt3 cDNA (Konishi et al., 1995) was reconstructed by PCR with a degenerate primer 5'- GCTCTCTAGATTATTC(T/C)CGTCC(A/G)CTTGCAGAGTAG-3' and the internal primer 5'-CAGGGCTCTTGATAAAG- GATCC-3' using rat brain phage cDNA library (lZAP, Stratagene) as a template (Brodbeck et al., 1999; Masure et al., 1999; Nakatani et al., 1999a). After con®rming the sequences, the wild-type and the myristylated version of Akt3 were subcloned into RCAS.S® Oncogene Neoplastic transformation by Akt2 and Akt3 I Mende et al 4421 a b Figure 3 Expression and in vitro kinase activity of the three Akt Figure 2 Transformation of CEF induced by Akt constructs. proteins. (a) Western blot analysis of the Akt proteins. Cells were Fertilized chicken eggs were obtained from SPAFAS. Primary lysed in Akt lysis buer supplemented with 1 mM microcystin CEF cultures, focus assays and DNA transfections using DMSO- (Calbiochem) (Aoki et al., 1998). Lysates containing 40 mgof polybrene method were described previously (Aoki et al., 1998). protein were separated by SDS ± PAGE and transferred to an Assay plates were evaluated after staining with crystal violet. Immobilon-P membrane (Millipore). The membrane was probed Tests for tumor formation in young chickens followed published with anti HA monoclonal antibody HA-11 (BabCo) followed by procedures (Aoki et al., 1998). Cell culture supernatants of horseradish peroxidase conjugated secondary antibody (Amer- transfected, RCAS construct-releasing CEF were serially diluted sham). Akt proteins were visualized by incubation with a and added to fresh secondary cultures of CEF in 35 mm plates. chemiluminescent substrate (Renaissance Plus, NEN). (b) Immune The cultures were overlaid with nutrient agar for 3 weeks and complex kinase assay. The Akt proteins were immunoprecipitated stained with crystal violet (Aoki et al., 2000). See Table 1 for the from the lysates containing 80 mg of protein with anti HA titer of individual constructs antibody and protein G sepharose beads (Pharmacia).
Recommended publications
  • Selective Targeting of Cyclin E1-Amplified High-Grade Serous Ovarian Cancer by Cyclin-Dependent Kinase 2 and AKT Inhibition
    Published OnlineFirst September 23, 2016; DOI: 10.1158/1078-0432.CCR-16-0620 Biology of Human Tumors Clinical Cancer Research Selective Targeting of Cyclin E1-Amplified High-Grade Serous Ovarian Cancer by Cyclin- Dependent Kinase 2 and AKT Inhibition George Au-Yeung1,2, Franziska Lang1, Walid J. Azar1, Chris Mitchell1, Kate E. Jarman3, Kurt Lackovic3,4, Diar Aziz5, Carleen Cullinane1,6, Richard B. Pearson1,2,7, Linda Mileshkin2,8, Danny Rischin2,8, Alison M. Karst9, Ronny Drapkin10, Dariush Etemadmoghadam1,2,5, and David D.L. Bowtell1,2,7,11 Abstract Purpose: Cyclin E1 (CCNE1) amplification is associated with Results: We validate CDK2 as a therapeutic target by demon- primary treatment resistance and poor outcome in high-grade strating selective sensitivity to gene suppression. However, we found serous ovarian cancer (HGSC). Here, we explore approaches to that dinaciclib did not trigger amplicon-dependent sensitivity in a target CCNE1-amplified cancers and potential strategies to over- panel of HGSC cell lines. A high-throughput compound screen come resistance to targeted agents. identified synergistic combinations in CCNE1-amplified HGSC, Experimental Design: To examine dependency on CDK2 in including dinaciclib and AKT inhibitors. Analysis of genomic data CCNE1-amplified HGSC, we utilized siRNA and conditional from TCGA demonstrated coamplification of CCNE1 and AKT2. shRNA gene suppression, and chemical inhibition using dina- Overexpression of Cyclin E1 and AKT isoforms, in addition to ciclib, a small-molecule CDK2 inhibitor. High-throughput mutant TP53, imparted malignant characteristics in untransformed compound screening was used to identify selective synergistic fallopian tube secretory cells, the dominant site of origin of HGSC.
    [Show full text]
  • Expressed Gene Fusions As Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer
    Published OnlineFirst December 14, 2017; DOI: 10.1158/2159-8290.CD-17-0535 RESEARCH ARTICLE Expressed Gene Fusions as Frequent Drivers of Poor Outcomes in Hormone Receptor–Positive Breast Cancer Karina J. Matissek1,2, Maristela L. Onozato3, Sheng Sun1,2, Zongli Zheng2,3,4, Andrew Schultz1, Jesse Lee3, Kristofer Patel1, Piiha-Lotta Jerevall2,3, Srinivas Vinod Saladi1,2, Allison Macleay3, Mehrad Tavallai1,2, Tanja Badovinac-Crnjevic5, Carlos Barrios6, Nuran Beşe7, Arlene Chan8, Yanin Chavarri-Guerra9, Marcio Debiasi6, Elif Demirdögen10, Ünal Egeli10, Sahsuvar Gökgöz10, Henry Gomez11, Pedro Liedke6, Ismet Tasdelen10, Sahsine Tolunay10, Gustavo Werutsky6, Jessica St. Louis1, Nora Horick12, Dianne M. Finkelstein2,12, Long Phi Le2,3, Aditya Bardia1,2, Paul E. Goss1,2, Dennis C. Sgroi2,3, A. John Iafrate2,3, and Leif W. Ellisen1,2 ABSTRACT We sought to uncover genetic drivers of hormone receptor–positive (HR+) breast cancer, using a targeted next-generation sequencing approach for detecting expressed gene rearrangements without prior knowledge of the fusion partners. We identified inter- genic fusions involving driver genes, including PIK3CA, AKT3, RAF1, and ESR1, in 14% (24/173) of unselected patients with advanced HR+ breast cancer. FISH confirmed the corresponding chromo- somal rearrangements in both primary and metastatic tumors. Expression of novel kinase fusions in nontransformed cells deregulates phosphoprotein signaling, cell proliferation, and survival in three- dimensional culture, whereas expression in HR+ breast cancer models modulates estrogen-dependent growth and confers hormonal therapy resistance in vitro and in vivo. Strikingly, shorter overall survival was observed in patients with rearrangement-positive versus rearrangement-negative tumors. Cor- respondingly, fusions were uncommon (<5%) among 300 patients presenting with primary HR+ breast cancer.
    [Show full text]
  • GSK3 and Its Interactions with the PI3K/AKT/Mtor Signalling Network
    Heriot-Watt University Research Gateway GSK3 and its interactions with the PI3K/AKT/mTOR signalling network Citation for published version: Hermida, MA, Kumar, JD & Leslie, NR 2017, 'GSK3 and its interactions with the PI3K/AKT/mTOR signalling network', Advances in Biological Regulation, vol. 65, pp. 5-15. https://doi.org/10.1016/j.jbior.2017.06.003 Digital Object Identifier (DOI): 10.1016/j.jbior.2017.06.003 Link: Link to publication record in Heriot-Watt Research Portal Document Version: Peer reviewed version Published In: Advances in Biological Regulation Publisher Rights Statement: © 2017 Elsevier B.V. General rights Copyright for the publications made accessible via Heriot-Watt Research Portal is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy Heriot-Watt University has made every reasonable effort to ensure that the content in Heriot-Watt Research Portal complies with UK legislation. If you believe that the public display of this file breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 27. Sep. 2021 Accepted Manuscript GSK3 and its interactions with the PI3K/AKT/mTOR signalling network Miguel A. Hermida, J. Dinesh Kumar, Nick R. Leslie PII: S2212-4926(17)30124-0 DOI: 10.1016/j.jbior.2017.06.003 Reference: JBIOR 180 To appear in: Advances in Biological Regulation Received Date: 13 June 2017 Accepted Date: 23 June 2017 Please cite this article as: Hermida MA, Dinesh Kumar J, Leslie NR, GSK3 and its interactions with the PI3K/AKT/mTOR signalling network, Advances in Biological Regulation (2017), doi: 10.1016/ j.jbior.2017.06.003.
    [Show full text]
  • Characterization of Akt Overexpression in Miapaca-2 Cells
    ANTICANCER RESEARCH 28 : 957-964 (2008) Characterization of Akt Overexpression in MiaPaCa-2 Cells: Prohibitin Is an Akt Substrate both In Vitro and in Cells EDWARD KYU-HO HAN, THOMAS MCGONIGAL, CHRIS BUTLER, VINCENT L. GIRANDA and YAN LUO Abbott Laboratories, Department of R47S, Cancer Research, Global Pharmaceutical Research Division, Abbott Park, IL 60064, U.S.A. Abstract. Akt (PKB) is a serine/threonine protein kinase that is activated by growth factors, cytokines and insulin. that plays an important role in the transduction of signals Activated PI-3K produces PI-3,4,5-P 3 and PI-3,4,-P 2 that affecting apoptosis, cell proliferation and survival. The Akt interact with Akt and recruit Akt from the cytosol to the gene is frequently hyperactivated in tumors and has been plasma membrane. Following membrane localization, Akt is shown to be amplified in a number of types of human phosphorylated at Thr-308 in the T-loop (kinase activation cancers. Furthermore, Akt activity is elevated in cell lines loop) and Ser-473 in the carboxy-terminal tail (hydrophobic with the mutated PTEN tumor suppressor gene. These studies motif). Thr-308 phosphorylation is required for Akt establish Akt as an attractive target for cancer therapy. To activation, while Ser-473 phosphorylation is only needed for determine the roles of Akt1, Akt2 and Akt3 in signal maximal activity. Thr-308 is phosphorylated by 3- transduction, constitutively active Akt1, Akt2 and Akt3 was phophoinositide dependent kinase (PDK1), which is ectopically overexpressed in human pancreatic MiaPaCa-2 activated by PI-3K lipid products (2). cells.
    [Show full text]
  • Cellular Processes Regulating Cytoskeletal
    CELLULAR PROCESSES REGULATING CYTOSKELETAL SIGNALING, INSULIN RESISTANCE AND CALCIUM SIGNALING ARE COMMONLY MISREGULATED IN TYPE 1 AND TYPE 2 MYOTONIC DYSTROPHY PATIENTS by NRIPESH PRASAD A DISSERTATION Submitted in partial fulfillment of the requirements for the Degree of Doctor of Philosophy in The Biotechnology Science and Engineering Program to The School of Graduate Studies of The University of Alabama in Huntsville HUNTSVILLE, ALABAMA 2014 ACKNOWLEDGEMENT The journey we take is supported by many others. Acknowledgement for a few might be just a trifle thing written on a piece of paper. Nevertheless, in the true essence, it gives us an opportunity to remember and express our feelings to those, whom we love, revere and share our secrets. Here I get a great chance to express my token of thanks to people who helped and supported me to complete this journey. It is my sublime duty to express my deepest sense of gratitude and veneration to my advisor Dr. Shawn E. Levy, Director, Genomics Service Lab, HudsonAlpha Institute for Biotechnology for his sincere and indelible inspiration, constant encouragement, constructive criticism, meticulous guidance, sustained interest, immense patience, and supportive nature throughout the investigation of the the research and preparation of this manuscript. I express my deepest sense of reverence and indebtedness to the esteemed members of my Advisory Committee, Dr(s). Joseph Ng, Debra Moriarity, Devin Absher, Gerg Cooper and Luis Cruz-Vera for their valuable suggestions and encouragement at various stages of my research and thesis writing. Sincere regards are due to, Dean, Graduate School, University of Alabama in Huntsville, Huntsville, AL, USA for providing all the necessary help and support.
    [Show full text]
  • Genomically Amplified Akt3 Activates DNA Repair Pathway and Promotes Glioma Progression
    Genomically amplified Akt3 activates DNA repair pathway and promotes glioma progression Kristen M. Turnera,1, Youting Suna,2, Ping Jia, Kirsi J. Granberga,b, Brady Bernardc, Limei Hua, David E. Cogdella, Xinhui Zhoua, Olli Yli-Harjab, Matti Nykterb, Ilya Shmulevichc, W. K. Alfred Yungd, Gregory N. Fullera, and Wei Zhanga,3 Departments of aPathology and dNeuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030; bDepartment of Signal Processing, Tampere University of Technology, Tampere, 33720, Finland; and cDepartment of Systems Biology, Institute for Systems Biology, Seattle, WA 98109 Edited* by Webster K. Cavenee, Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA, and approved February 13, 2015 (received for review July 30, 2014) Akt is a robust oncogene that plays key roles in the development disease progression and maintenance, whereas Akt1 does not play and progression of many cancers, including glioma. We evaluated a major role (15, 16). Another study in transformed mouse astro- the differential propensities of the Akt isoforms toward progression cytes indicate that Akt isoform function is potentially modulated by in the well-characterized RCAS/Ntv-a mouse model of PDGFB-driven the genetic landscape of the tumor (17). The underlying basis be- low grade glioma. A constitutively active myristoylated form of hind these differences and the major pathways that the isoforms Akt1 did not induce high-grade glioma (HGG). In stark contrast, Akt2 use remains largely unknown. To our knowledge, our study is the and Akt3 showed strong progression potential with 78% and 97% first to use a glial-specific transgenic mouse model to demonstrate of tumors diagnosed as HGG, respectively.
    [Show full text]
  • Genetic Background of Ataxia in Children Younger Than 5 Years in Finland E444
    Volume 6, Number 4, August 2020 Neurology.org/NG A peer-reviewed clinical and translational neurology open access journal ARTICLE Genetic background of ataxia in children younger than 5 years in Finland e444 ARTICLE Cerebral arteriopathy associated with heterozygous variants in the casitas B-lineage lymphoma gene e448 ARTICLE Somatic SLC35A2 mosaicism correlates with clinical fi ndings in epilepsy brain tissuee460 ARTICLE Synonymous variants associated with Alzheimer disease in multiplex families e450 Academy Officers Neurology® is a registered trademark of the American Academy of Neurology (registration valid in the United States). James C. Stevens, MD, FAAN, President Neurology® Genetics (eISSN 2376-7839) is an open access journal published Orly Avitzur, MD, MBA, FAAN, President Elect online for the American Academy of Neurology, 201 Chicago Avenue, Ann H. Tilton, MD, FAAN, Vice President Minneapolis, MN 55415, by Wolters Kluwer Health, Inc. at 14700 Citicorp Drive, Bldg. 3, Hagerstown, MD 21742. Business offices are located at Two Carlayne E. Jackson, MD, FAAN, Secretary Commerce Square, 2001 Market Street, Philadelphia, PA 19103. Production offices are located at 351 West Camden Street, Baltimore, MD 21201-2436. Janis M. Miyasaki, MD, MEd, FRCPC, FAAN, Treasurer © 2020 American Academy of Neurology. Ralph L. Sacco, MD, MS, FAAN, Past President Neurology® Genetics is an official journal of the American Academy of Neurology. Journal website: Neurology.org/ng, AAN website: AAN.com CEO, American Academy of Neurology Copyright and Permission Information: Please go to the journal website (www.neurology.org/ng) and click the Permissions tab for the relevant Mary E. Post, MBA, CAE article. Alternatively, send an email to [email protected].
    [Show full text]
  • Androgen Receptor
    RALTITREXED Dihydrofolate reductase BORTEZOMIB IsocitrateCannabinoid dehydrogenase CB1EPIRUBICIN receptor HYDROCHLORIDE [NADP] cytoplasmic VINCRISTINE SULFATE Hypoxia-inducible factor 1 alpha DOXORUBICINAtaxin-2 HYDROCHLORIDENIFENAZONEFOLIC ACID PYRIMETHAMINECellular tumor antigen p53 Muscleblind-likeThyroidVINBURNINEVINBLASTINETRIFLURIDINE protein stimulating 1 DEQUALINIUM SULFATEhormone receptor CHLORIDE Menin/Histone-lysine N-methyltransferasePHENELZINE MLLLANATOSIDE SULFATE C MELATONINDAUNORUBICINBETAMETHASONEGlucagon-like HYDROCHLORIDEEndonuclease peptide 4 1 receptor NICLOSAMIDEDIGITOXINIRINOTECAN HYDROCHLORIDE HYDRATE BISACODYL METHOTREXATEPaired boxAZITHROMYCIN protein Pax-8 ATPase family AAA domain-containing proteinLIPOIC 5 ACID, ALPHA Nuclear receptorCLADRIBINEDIGOXIN ROR-gammaTRIAMTERENE CARMUSTINEEndoplasmic reticulum-associatedFLUOROURACIL amyloid beta-peptide-binding protein OXYPHENBUTAZONEORLISTAT IDARUBICIN HYDROCHLORIDE 6-phospho-1-fructokinaseHeat shockSIMVASTATIN protein beta-1 TOPOTECAN HYDROCHLORIDE AZACITIDINEBloom syndromeNITAZOXANIDE protein Huntingtin Human immunodeficiency virus typeTIPRANAVIR 1 protease VitaminCOLCHICINE D receptorVITAMIN E FLOXURIDINE TAR DNA-binding protein 43 BROMOCRIPTINE MESYLATEPACLITAXEL CARFILZOMIBAnthrax lethalFlap factorendonucleasePrelamin-A/C 1 CYTARABINE Vasopressin V2 receptor AMITRIPTYLINEMicrotubule-associated HYDROCHLORIDERetinoidTRIMETHOPRIM proteinMothers X receptor tau against alpha decapentaplegic homolog 3 Histone-lysine N-methyltransferase-PODOFILOX H3 lysine-9OXYQUINOLINE
    [Show full text]
  • Targeting BRAF for Patients with Melanoma
    British Journal of Cancer (2011) 104, 392 – 398 & 2011 Cancer Research UK All rights reserved 0007 – 0920/11 www.bjcancer.com Minireview Targeting BRAF for patients with melanoma ,1,3 2 2,3 H-T Arkenau* , R Kefford and GV Long 1Sarah Cannon Research UK, 93 Harley Street, London W1G 6AD, UK; 2Melanoma Institute Australia and Westmead Hospital, University of Sydney, New South Wales, Australia The prognosis of patients with metastatic melanoma is poor and not influenced by systemic therapy with cytotoxic drugs. New targeted agents directed against the RAS-RAF-MEK-ERK pathway show promising activity in early clinical development and particular interest is focused on selective inhibitors of mutant BRAF, which is present in one half of the cases of metastatic melanoma. The majority of patients on early trials of these drugs develop secondary resistance and subsequent disease progression and it is, therefore, critical to understand the underlying escape mechanisms leading to resistance. British Journal of Cancer (2011) 104, 392–398. doi:10.1038/sj.bjc.6606030 www.bjcancer.com Published online 7 December 2010 & 2011 Cancer Research UK Keywords: BRAF; BRAFV600E; melanoma; PI3K-AKT-mTOR; RAS-RAF-MEK-ERK MELANOMA: A HETEROGENEOUS DISEASE website). The mutations are either in the activating segment in exon 15 or the glycine-rich loop (P-loop) in exon 11 of the kinase Metastases develop in 10–15% of patients with cutaneous domain of the BRAF protein (Figure 2). The point mutation in melanoma and there is no evidence from phase-III trials that DNA (1799T-A) resulting in a single amino-acid substitution at systemic treatment prolongs survival, nor is there an effective Valine 600 to Glutamic acid in the activating segment (V600E, adjuvant therapy after resection of high risk Stage I–III disease previously known as V599E) is the most common change, and is (Thompson et al, 2005).
    [Show full text]
  • Mtorc1 Is Necessary but Mtorc2 and Gsk3b Are Inhibitory for AKT3
    RESEARCH ARTICLE mTORC1 is necessary but mTORC2 and GSK3b are inhibitory for AKT3-induced axon regeneration in the central nervous system Linqing Miao1†, Liu Yang1†, Haoliang Huang1, Feisi Liang1, Chen Ling2, Yang Hu1,3* 1Shriners Hospitals Pediatric Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, United States; 2Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, United States; 3Department of Anatomy and Cell Biology, Temple University Lewis Katz School of Medicine, Philadelphia, United States Abstract Injured mature CNS axons do not regenerate in mammals. Deletion of PTEN, the negative regulator of PI3K, induces CNS axon regeneration through the activation of PI3K-mTOR signaling. We have conducted an extensive molecular dissection of the cross-regulating mechanisms in axon regeneration that involve the downstream effectors of PI3K, AKT and the two mTOR complexes (mTORC1 and mTORC2). We found that the predominant AKT isoform in CNS, AKT3, induces much more robust axon regeneration than AKT1 and that activation of mTORC1 and inhibition of GSK3b are two critical parallel pathways for AKT-induced axon regeneration. Surprisingly, phosphorylation of T308 and S473 of AKT play opposite roles in GSK3b phosphorylation and inhibition, by which mTORC2 and pAKT-S473 negatively regulate axon regeneration. Thus, our study revealed a complex neuron-intrinsic balancing mechanism involving *For correspondence: yanghu@ AKT as the nodal point of PI3K, mTORC1/2 and GSK3b that coordinates both positive and negative temple.edu cues to regulate adult CNS axon regeneration. † These authors contributed DOI: 10.7554/eLife.14908.001 equally to this work Competing interests: The authors declare that no competing interests exist.
    [Show full text]
  • Inhibition of ERK 1/2 Kinases Prevents Tendon Matrix Breakdown Ulrich Blache1,2,3, Stefania L
    www.nature.com/scientificreports OPEN Inhibition of ERK 1/2 kinases prevents tendon matrix breakdown Ulrich Blache1,2,3, Stefania L. Wunderli1,2,3, Amro A. Hussien1,2, Tino Stauber1,2, Gabriel Flückiger1,2, Maja Bollhalder1,2, Barbara Niederöst1,2, Sandro F. Fucentese1 & Jess G. Snedeker1,2* Tendon extracellular matrix (ECM) mechanical unloading results in tissue degradation and breakdown, with niche-dependent cellular stress directing proteolytic degradation of tendon. Here, we show that the extracellular-signal regulated kinase (ERK) pathway is central in tendon degradation of load-deprived tissue explants. We show that ERK 1/2 are highly phosphorylated in mechanically unloaded tendon fascicles in a vascular niche-dependent manner. Pharmacological inhibition of ERK 1/2 abolishes the induction of ECM catabolic gene expression (MMPs) and fully prevents loss of mechanical properties. Moreover, ERK 1/2 inhibition in unloaded tendon fascicles suppresses features of pathological tissue remodeling such as collagen type 3 matrix switch and the induction of the pro-fbrotic cytokine interleukin 11. This work demonstrates ERK signaling as a central checkpoint to trigger tendon matrix degradation and remodeling using load-deprived tissue explants. Tendon is a musculoskeletal tissue that transmits muscle force to bone. To accomplish its biomechanical function, tendon tissues adopt a specialized extracellular matrix (ECM) structure1. Te load-bearing tendon compart- ment consists of highly aligned collagen-rich fascicles that are interspersed with tendon stromal cells. Tendon is a mechanosensitive tissue whereby physiological mechanical loading is vital for maintaining tendon archi- tecture and homeostasis2. Mechanical unloading of the tissue, for instance following tendon rupture or more localized micro trauma, leads to proteolytic breakdown of the tissue with severe deterioration of both structural and mechanical properties3–5.
    [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]