17 research outputs found

    The Receptor Tyrosine Kinase Alk Controls Neurofibromin Functions in Drosophila Growth and Learning

    Get PDF
    Anaplastic Lymphoma Kinase (Alk) is a Receptor Tyrosine Kinase (RTK) activated in several cancers, but with largely unknown physiological functions. We report two unexpected roles for the Drosophila ortholog dAlk, in body size determination and associative learning. Remarkably, reducing neuronal dAlk activity increased body size and enhanced associative learning, suggesting that its activation is inhibitory in both processes. Consistently, dAlk activation reduced body size and caused learning deficits resembling phenotypes of null mutations in dNf1, the Ras GTPase Activating Protein-encoding conserved ortholog of the Neurofibromatosis type 1 (NF1) disease gene. We show that dAlk and dNf1 co-localize extensively and interact functionally in the nervous system. Importantly, genetic or pharmacological inhibition of dAlk rescued the reduced body size, adult learning deficits, and Extracellular-Regulated-Kinase (ERK) overactivation dNf1 mutant phenotypes. These results identify dAlk as an upstream activator of dNf1-regulated Ras signaling responsible for several dNf1 defects, and they implicate human Alk as a potential therapeutic target in NF1

    Deficiency screen summary.

    No full text
    <p>Indicated are the number of chromosome 1, 2L and 2R deficiencies screened, the fraction of genes uncovered (based on the FB2013_03 FlyBase release), the number of <i>dNf1</i> modifying deficiencies and loci identified, and the number of non-specific modifiers.</p

    Validation of <i>dNf1</i> modifiers with neuronal functions.

    No full text
    <p>(A) <i>Ras2-Gal4</i> or <i>C23-Gal4</i> driven neuronal RNAi knockdown of <i>CCKLR-17D1</i> but not <i>CCKLR-17D3</i> suppressed the <i>dNf1</i> pupal size defect. (B) Identification of dynamin-associated protein 160 (Dap160) as a suppressor of <i>dNf1</i> growth. Neuronal RNAi targeting of <i>Dap160</i> increased <i>dNf1</i> pupal size as did two <i>Dap160</i> loss-of-function alleles. (C) Two <i>elav</i> alleles dominantly suppress the <i>dNf1</i> size defect. (D) Neuronal expression of a Rab9 RNAi transgene or of a dominant negative Rab9 mutant suppresses the <i>dNf1</i> size defect.</p

    Deficiency screen for dominant modifiers of the <i>dNf1</i> growth defect.

    No full text
    <p>Isogenic 1<sup>st</sup> and 2<sup>nd</sup> chromosomes deficiencies from the Exelixis, DrosDel and Bloomington Stock Center collections were tested for their ability to alter <i>dNf1</i> female pupal size. Crossing schemes to generate <i>Df(1)/+; dNf1<sup>E2</sup></i> (A) and <i>Df(2)/CyO; dNf1<sup>E2</sup></i> (B) screening stocks. The <i>tubby</i>-marked <i>TM6B</i> 3<sup>rd</sup> chromosome balancer allowed the selection of <i>dNf1<sup>E2</sup></i> homozygotes for measurements. (C) Examples of deficiencies that suppress or enhance the <i>dNf1</i> size defect. Scale bar = 1 mm.</p

    Restoration of systemic growth by <i>dNf1</i> and cAMP/PKA involves different tissues.

    No full text
    <p><i>Act5C</i>-<i>Gal4</i> driven ubiquitous <i>dNf1</i> re-expression, or <i>elav</i>-<i>Gal4</i> and <i>Ras2</i>-<i>Gal4</i> driven neuronal re-expression rescues the <i>dNf1</i> pupal size defect, whereas <i>dnc</i> RNAi or UAS-PKA* expression controlled by the same drivers is ineffective. By contrast, expressing <i>dNf1</i> in specific parts of the neuroendocrine ring gland with the <i>Akh</i>-<i>Gal4</i>, Feb36-<i>Gal4</i> or Aug21-<i>Gal4</i> drivers fails to rescue, whereas using the same drivers to express <i>dnc</i> RNAi or attenuated UAS-PKA* transgenes does increase <i>dNf1</i> pupal size. All crosses produced viable adults unless otherwise indicated.</p>†<p>denotes lethality, SV sub-viable, n/a not applicable, NR non-rescue.</p><p>The data shown summarize results of a larger effort to identify the tissues in which <i>dNf1</i> and cAMP/PKA affect systemic growth. Full results are shown in <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1003958#pgen.1003958.s013" target="_blank">Table S5</a>.</p

    <i>dNf1</i> systemic growth related RAS/ERK and cAMP/PKA signals appear functionally and topographically distinct.

    No full text
    <p>(A) The elevated larval CNS pERK level of <i>dNf1</i> mutants is reduced by neuronal expression of <i>dNf1</i>, but not by neuronal or heat-shock induced ubiquitous expression of PKA*. Western blot of pERK levels in larval CNS of the indicated genotypes. In lane 6, larvae received a daily 20 min 37°C heat shock throughout development, a protocol that suppresses the <i>dNf1</i> growth defect <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1003958#pgen.1003958-The1" target="_blank">[4]</a>. (B) Structure of <i>UAS-PKA*</i> transgenes with 1 to 5 UAS elements. The lethality of these transgenes when driven with either <i>Ac5C-Gal4</i> or <i>elav-Gal4</i> is indicated by † whereas (−) indicates viable offspring. (C) Western blot of adult head lysates showing relative expression of <i>GMR-Gal4</i>-driven transgenic PKA*. Tubulin is used as a loading control. (D) Expression of PKA* or knockdown of <i>dnc</i> by shRNAi in the ring gland rescues the <i>dNf1</i> pupal size defect. In contrast, <i>UAS-dNf1</i> expression with the same ring gland drivers fails to restore systemic growth. (E–H) Expression pattern of <i>Akh-Gal4</i> driving <i>UAS-GFP</i>, co-stained with DAPI and anti-dNF1. GFP expression in the corpora cardiaca (CC) is indicated. Scale bar = 50 µm. As previously noted <a href="http://www.plosgenetics.org/article/info:doi/10.1371/journal.pgen.1003958#pgen.1003958-Walker2" target="_blank">[74]</a>, anti-dNf1 staining is strong in the CNS, whereas staining in the ring gland is close to background.</p

    Modifying deficiencies and identification of responsible genes.

    No full text
    <p>Modifying deficiencies for which the responsible <i>dNf1</i> interacting gene has been identified. The cytological location, and the dominant effect on <i>dNf1</i> pupal size (SUP - suppressor, ENH – enhancer) of each deficiency is given. The responsible genes for each modifying deficiency are shown with the mutant alleles, VDRC and TRiP RNAi lines used in their identification. Expression of RNAi transgenes was induced with the <i>Ras2</i>-<i>Gal4</i>, <i>elav</i>-<i>Gal4</i>, <i>n-syb</i>-<i>Gal4</i> and/or C23-<i>Gal4</i> drivers. Abbreviations: hypo: hypomorphic; leth: lethal; lof: loss-of-function; amorph: amorphic; Δ: deletion; via: viable.</p

    Identification of modifying genes with undetermined roles in <i>dNf1</i> suppression.

    No full text
    <p>(A) Validation of <i>NAAT1</i>, <i>HERC2</i> and <i>Hs3st-B</i> as <i>dNf1</i> modifiers. All three genes were identified by systematic RNAi screening of genes uncovered by suppressing deficiencies. (B) Loss-of-function alleles of Class C Vacuolar Protein Sorting complex subunits <i>carnation</i> (<i>car/Vps33A</i>) and <i>deep-orange</i> (<i>dor/Vps18</i>) increase <i>dNf1</i> pupal size. C) RNAi-mediated neuronal <i>car</i> or <i>dor</i> knockdown was not particularly effective, suggesting these genes may function elsewhere to modify <i>dNf1</i>-dependent growth.</p
    corecore