Mediated Sorting of INPP5E Into the Cilium Is Determined by Cargo-Carrier Affinity

Total Page:16

File Type:pdf, Size:1020Kb

Mediated Sorting of INPP5E Into the Cilium Is Determined by Cargo-Carrier Affinity ARTICLE Received 2 Sep 2015 | Accepted 18 Mar 2016 | Published 11 Apr 2016 DOI: 10.1038/ncomms11366 OPEN PDE6d-mediated sorting of INPP5E into the cilium is determined by cargo-carrier affinity Eyad Kalawy Fansa1,*, Stefanie Kristine Ko¨sling1,*, Eldar Zent1, Alfred Wittinghofer1 & Shehab Ismail2 The phosphodiesterase 6 delta subunit (PDE6d) shuttles several farnesylated cargos between membranes. The cargo sorting mechanism between cilia and other compartments is not understood. Here we show using the inositol polyphosphate 50-phosphatase E (INPP5E) and the GTP-binding protein (Rheb) that cargo sorting depends on the affinity towards PDE6d and the specificity of cargo release. High-affinity cargo is exclusively released by the ciliary transport regulator Arl3, while low-affinity cargo is released by Arl3 and its non-ciliary homologue Arl2. Structures of PDE6d/cargo complexes reveal the molecular basis of the sorting signal which depends on the residues at the À 1 and À 3 positions relative to far- nesylated cysteine. Structure-guided mutation allows the generation of a low-affinity INPP5E mutant which loses exclusive ciliary localization. We postulate that the affinity to PDE6d and the release by Arl2/3 in addition to a retention signal are the determinants for cargo sorting and enrichment at its destination. 1 Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany. 2 CR-UK Beatson Institute, Garscube Estate Switchback Road, Glasgow G61 1BD, UK. * These authors contributed equally to this work. Correspondence and requests for materials should be addressed to A.W. (email: [email protected]) or to S.I. (email: [email protected]). NATURE COMMUNICATIONS | 7:11366 | DOI: 10.1038/ncomms11366 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/ncomms11366 rimary cilia are antenna-like microtubule-based cell surface the import into cilia is dependent on the carrier proteins PDE6d, protrusions which can be found on eukaryotic cells and UNC119a and UNC119b and on Arl3 as displacement Pserve as sensory organelles. Genetic disorders affecting factor13,25,28,30,34. However, it has been extensively documented structure or function of cilia result in a large number of diseases that Ras proteins as well as Rheb require PDE6d for their proper collectively termed ciliopathies1,2. While the cilium appears as a localization at the plasma membrane or internal membranes, but protrusion in the plasma membrane that is open to the cell body, do not appear to be localized in cilia15,16. the ciliary content and membrane composition are different than This begs the question about the mechanism of PDE6d- that of the cell body and plasma membrane3,4. This is in part mediated sorting of farnesylated cargo between the cilium and achieved by the presence of a diffusion and transport barrier, other cellular compartments. Thus, we set out to investigate the where entry and exit decisions of ciliary components have to be molecular basis of farnesylated cargo sorting using ciliary INPP5E taken5,6. and non-ciliary Rheb as an example. Here, we show that a PDE6d is a prenyl-binding protein that was originally 100-fold difference in the binding affinity of farnesylated cargo discovered as the delta subunit of rod photoreceptor-specific with PDE6d and the specific release of high-affinity cargo by phosphodiesterase PDE6 (ref. 7). It was found as a solubilizing activated Arl3GTP determines cargo sorting into cilia, while factor for the prenylated subunits of this enzyme and was later low-affinity cargo can be released by both Arl3GTP and shown to be a general prenyl-binding protein (hence also called Arl2GTP and stays outside the cilium. Moreover, we show by PrBP/PDE6d)8–11. PDE6d was shown to bind prenylated peptides structural, biochemical and cell biological approaches, how and or proteins of the Ras subfamily with approximately micromolar why the binding affinity is dependent on the residues at the À 1 affinity12,13 and to play a critical role in their cellular and À 3 positions preceding the farnesylated cysteine and that distribution14–16. Since it is believed to be crucial for the sorting of farnesylated cargo can be manipulated by changing the localization and thus the activity of the oncoprotein Ras, affinity to PDE6d. inhibitors of the Ras-PDE6d complex were actually considered 17 as promising Ras drug candidates . Results INPP5E belongs to the inositol polyphosphate 50-phosphatase INPP5E and Rheb localization and binding affinity to PDE6d. family that hydrolyzes the 50-phosphate of phosphatidylinositols 18,19 Using IMCD3 cells stably expressing either INPP5E or Rheb fused and localizes to primary cilia . The importance of the 35 0 to a localization and tandem affinity purification (LAP) tag ,we 5 -phosphatase activity for ciliary function is underscored by can show that INPP5E localizes almost exclusively to the primary the finding that INPP5E is mutated in Joubert syndrome, a 18–20 cilium with very small fraction in the cell body (Fig. 1a; upper), ciliopathy characterized by motor and intellectual disabilities , which is consistent with previous reports18,19,26. In contrast, Rheb and that the gene mutated in the OCRL (Oculocerebrorenal) or mainly localizes to endomembranes (Fig. 1a; lower), this Lowe syndrome also encodes an inositol polyphosphate observation is consistent with previous reports13,36. Given that 50-phosphatase21,22. INPP5E contains a C-terminal CaaX motif 23 the prenyl-binding protein PDE6d is the shuttle factor mediating where the C-terminal residue Cys644 is farnesylated . A mutation the localization of INPP5E and Rheb13,16,18,25,26, we set out to encoding a stop codon near to the CaaX motif (Q627) of INPP5E 18 characterize the interaction of PDE6d with INPP5E and Rheb. was identified in a family with MORM syndrome , a ciliopathy Previously we have shown that farnesylated C-terminal peptides characterized by intellectual disability, obesity, retinal dystrophy derived from Rheb or KRas bind to PDE6d in exactly the same and micropenis24. This mutation was shown to affect INPP5E 25 way and with similar affinities as the full-length farnesylated ciliary localization, which in combination with other reports proteins12,13. Hence, we used a fluorescently labelled C-terminal indicates the importance of the C-terminus and its farnesylation farnesylated and carboxy-methylated peptide of INPP5E (residues for the ciliary localization of INPP5E (ref. 18). 637–644) and Rheb (residues 175–181) to measure the affinity to Recently, PDE6d was co-purified with INPP5E and siRNA- PDE6d by fluorescence polarization. Figure 1b (left) shows that d mediated knockdown of PDE6 resulted in impaired ciliary PDE6d binds to INPP5E peptide with low nanomolar affinity localization of INPP5E (ref. 26). Moreover, a PDE6d deletion (Kd ¼ 3.7 nM±0.2,±indicates s.d., n ¼ 9). In contrast, the affinity mutation, which was identified in Joubert syndrome, was between PDE6d and the farnesylated C-terminal peptide of Rheb shown to impair the targeting of farnesylated INPP5E protein falls into the submicromolar range (K ¼ 445±83 nM,±indicates 25 d d to the primary cilium . Knockdown of PDE6 also impeded the s.d., n ¼ 10) (Fig. 1b; right), which is in the same range with the transport of GRK1 and PDE6 catalytic subunits to photoreceptor previously described values12,13. These data raised the question, outer segments, which are considered specialized forms of whether the almost 100-fold higher affinity of INPP5E towards cilia27,28. PDE6d as compared to Rheb is involved in the sorting The homologous small Arf-like GTP-binding proteins Arl2 mechanism of these two proteins to different destinations. and Arl3 have been shown to act as nucleotide-dependent-specific release factors of farnesylated cargo from PDE6d in vitro and in vivo. Structural and kinetic analyses have shown that Arl2/3 act High-affinity cargo is specifically released by Arl3GTP. allosterically to increase the dissociation rate constants for cargo- Towards an explanation for the possible sorting mechanism that carrier complexes13,15,29,30. In contrast, it was shown recently by leaves some PDE6d-cargo in the cell body but allows others to be pull-down experiments with cellular extracts that Arl3 but enriched in the cilia we turned to the release activities of Arl2 and not Arl2 can efficiently release INPP5E from its complex with Arl3. Both GTP-binding proteins in their active conformation PDE6d (ref. 25). have been shown to be responsible for releasing cargo from In analogy to nuclear localization signals a number of different PDE6d. While Arl2 is a non-ciliary protein, Arl3 localizes along ciliary localization signals have been identified for different the length of the cilium37. Using fluorescence polarization, we transmembrane proteins31–33. However, not much is known measured the release of INPP5E and Rheb peptides from PDE6d about the molecular mechanism of how these signals are by the addition of Arl2 or Arl3 bound to the non-hydrolysable recognized and how decisions on ciliary entry based on these GTP analogue GppNHp. The data show that Rheb peptide can be signals are made. For certain membrane-associated, post- released by both Arl2GppNHp and Arl3GppNHp (Fig. 2a), translationally modified proteins carrying an N-terminal supporting earlier observations13. In contrast, INPP5E peptide myristoyl or a C-terminal prenyl motif, it has been shown that can only be released by Arl3GppNHp under the same conditions 2 NATURE COMMUNICATIONS | 7:11366 | DOI:
Recommended publications
  • Regulation of Phosphoinositide Levels in the Retina by Protein Tyrosine Phosphatase 1B and Growth Factor Receptor-Bound Protein 14
    biomolecules Article Regulation of Phosphoinositide Levels in the Retina by Protein Tyrosine Phosphatase 1B and Growth Factor Receptor-Bound Protein 14 Raju V. S. Rajala 1,2,3,4,* , Austin McCauley 1,4, Rahul Rajala 3,5 , Kenneth Teel 1,4 and Ammaji Rajala 1,4 1 Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; [email protected] (A.M.); [email protected] (K.T.); [email protected] (A.R.) 2 Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA 3 Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; [email protected] 4 Dean McGee Eye Institute, Oklahoma City, OK 73104, USA 5 Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA * Correspondence: [email protected]; Tel.: +1-405-271-8255; Fax: +1-405-271-8128 Abstract: Protein tyrosine kinases and protein phosphatases play a critical role in cellular regulation. The length of a cellular response depends on the interplay between activating protein kinases and deactivating protein phosphatases. Protein tyrosine phosphatase 1B (PTP1B) and growth factor receptor-bound protein 14 (Grb14) are negative regulators of receptor tyrosine kinases. However, in the retina, we have previously shown that PTP1B inactivates insulin receptor signaling, whereas phosphorylated Grb14 inhibits PTP1B activity. In silico docking of phosphorylated Grb14 and PTP1B Citation: Rajala, R.V.S.; McCauley, indicate critical residues in PTP1B that may mediate the interaction. Phosphoinositides (PIPs) are A.; Rajala, R.; Teel, K.; Rajala, A. acidic lipids and minor constituents in the cell that play an important role in cellular processes.
    [Show full text]
  • The Role of Genetic Variation in Predisposition to Alcohol-Related Chronic Pancreatitis
    The Role of Genetic Variation in Predisposition to Alcohol-related Chronic Pancreatitis Thesis submitted in accordance with the requirements of the University of Liverpool for the degree of Doctor in Philosophy by Marianne Lucy Johnstone April 2015 The Role of Genetic Variation in Predisposition to Alcohol-related Chronic Pancreatitis 2015 Abstract Background Chronic pancreatitis (CP) is a disease of fibrosis of the pancreas for which alcohol is the main causative agent. However, only a small proportion of alcoholics develop chronic pancreatitis. Genetic polymorphism may affect pancreatitis risk. Aim To determine the factors required to classify a chronic pancreatic population and identify genetic variations that may explain why only some alcoholics develop chronic pancreatitis. Methods The most appropriate method of diagnosing CP was assessed using a systematic review. Genetics of different populations of alcohol-related chronic pancreatitics (ACP) were explored using four different techniques: genome-wide association study (GWAS); custom arrays; PCR of variable nucleotide tandem repeats (VNTR) and next generation sequencing (NGS) of selected genes. Results EUS and sMR were identified as giving the overall best sensitivity and specificity for diagnosing CP. GWAS revealed two associations with CP (identified and replicated) at PRSS1-PRSS2_rs10273639 (OR 0.73, 95% CI 0.68-0.79) and X-linked CLDN2_rs12688220 (OR 1.39, 1.28-1.49) and the association was more pronounced in the ACP group (OR 0.56, 0.48-0.64)and OR 2.11, 1.84-2.42). The previously identified VNTR in CEL was shown to have a lower frequency of the normal repeat in ACP than alcoholic liver disease (ALD; OR 0.61, 0.41-0.93).
    [Show full text]
  • Whole Exome Sequencing Analyses Reveal Gene–Microbiota Interactions
    Inflammatory bowel disease ORIGINAL RESEARCH Whole exome sequencing analyses reveal gene– Gut: first published as 10.1136/gutjnl-2019-319706 on 10 July 2020. Downloaded from microbiota interactions in the context of IBD Shixian Hu ,1,2 Arnau Vich Vila ,1,2 Ranko Gacesa,1,2 Valerie Collij,1,2 Christine Stevens,3 Jack M Fu,4,5,6 Isaac Wong,4,5 Michael E Talkowski,4,5,6,7,8 Manuel A Rivas,9 Floris Imhann,1,2 Laura Bolte,1,2 Hendrik van Dullemen,1 Gerard Dijkstra ,1 Marijn C Visschedijk,1 Eleonora A Festen,1 Ramnik J Xavier,10,11 Jingyuan Fu,2,12 Mark J Daly,3 Cisca Wijmenga,2 Alexandra Zhernakova,2 Alexander Kurilshikov,2 Rinse K Weersma 1 ► Additional material is ABSTRact published online only. To view Objective Both the gut microbiome and host genetics Significance of this study please visit the journal online are known to play significant roles in the pathogenesis (http:// dx. doi. org/ 10. 1136/ What is already known about this subject? gutjnl- 2019- 319706). of IBD. However, the interaction between these two factors and its implications in the aetiology of IBD remain ► Gene–microbiome interactions are important in For numbered affiliations see the pathogenesis of IBD. end of article. underexplored. Here, we report on the influence of host genetics on the gut microbiome in IBD. ► Multiple genetic and epidemiological factors have been identified to be associated to Correspondence to Design To evaluate the impact of host genetics on Professor Rinse K Weersma; the gut microbiota of patients with IBD, we combined changes in gut microbiome homeostasis in both r.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Epigenomic Signatures in Liver and Blood of Wilson Disease Patients Include Hypermethylation of Liver‑Specifc Enhancers Charles E
    Mordaunt et al. Epigenetics & Chromatin (2019) 12:10 https://doi.org/10.1186/s13072-019-0255-z Epigenetics & Chromatin RESEARCH Open Access Epigenomic signatures in liver and blood of Wilson disease patients include hypermethylation of liver-specifc enhancers Charles E. Mordaunt1, Dorothy A. Kiefer2, Noreene M. Shibata2, Anna Członkowska3, Tomasz Litwin3, Karl‑Heinz Weiss4, Yihui Zhu1, Christopher L. Bowlus2, Souvik Sarkar2, Stewart Cooper5, Yu‑Jui Yvonne Wan6, Mohamed R. Ali7, Janine M. LaSalle1† and Valentina Medici2*† Abstract Background: Wilson disease (WD) is an autosomal recessive disease caused by mutations in ATP7B encoding a copper transporter. Consequent copper accumulation results in a variable WD clinical phenotype involving hepatic, neurologic, and psychiatric symptoms, without clear genotype–phenotype correlations. The goal of this study was to analyze alterations in DNA methylation at the whole‑genome level in liver and blood from patients with WD to investigate epigenomic alterations associated with WD diagnosis and phenotype. We used whole‑genome bisulfte sequencing (WGBS) to examine distinct cohorts of WD subjects to determine whether DNA methylation could dif‑ ferentiate patients from healthy subjects and subjects with other liver diseases and distinguish between diferent WD phenotypes. Results: WGBS analyses in liver identifed 969 hypermethylated and 871 hypomethylated diferentially methyl‑ ated regions (DMRs) specifcally identifying patients with WD, including 18 regions with genome‑wide signifcance. WD‑specifc liver DMRs were associated with genes enriched for functions in folate and lipid metabolism and acute infammatory response and could diferentiate early from advanced fbrosis in WD patients. Functional annotation revealed that WD‑hypermethylated liver DMRs were enriched in liver‑specifc enhancers, fanking active liver pro‑ moters, and binding sites of liver developmental transcription factors, including Hepatocyte Nuclear Factor 4 alpha (HNF4A), Retinoid X Receptor alpha (RXRA), Forkhead Box A1 (FOXA1), and FOXA2.
    [Show full text]
  • A Transient Role of the Ciliary Gene Inpp5e in Controlling Direct Versus
    RESEARCH ARTICLE A transient role of the ciliary gene Inpp5e in controlling direct versus indirect neurogenesis in cortical development Kerstin Hasenpusch-Theil1,2, Christine Laclef3†, Matt Colligan1, Eamon Fitzgerald1, Katherine Howe1, Emily Carroll1, Shaun R Abrams4, Jeremy F Reiter4,5, Sylvie Schneider-Maunoury3, Thomas Theil1,2* 1Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; 2Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, United Kingdom; 3Sorbonne Universite´, CNRS UMR7622, INSERM U1156, Institut de Biologie Paris Seine (IBPS) - Developmental Biology Unit, Paris, France; 4Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States; 5Chan Zuckerberg Biohub, San Francisco, United States Abstract During the development of the cerebral cortex, neurons are generated directly from radial glial cells or indirectly via basal progenitors. The balance between these division modes determines the number and types of neurons formed in the cortex thereby affecting cortical functioning. Here, we investigate the role of primary cilia in controlling the decision between forming neurons directly or indirectly. We show that a mutation in the ciliary gene Inpp5e leads to a transient increase in direct neurogenesis and subsequently to an overproduction of layer V *For correspondence: neurons in newborn mice. Loss of Inpp5e also affects ciliary structure coinciding with reduced Gli3 [email protected] repressor levels. Genetically restoring Gli3 repressor rescues the decreased indirect neurogenesis Present address: †Sorbonne in Inpp5e mutants. Overall, our analyses reveal how primary cilia determine neuronal subtype Universite´, Institut du Fer a` composition of the cortex by controlling direct versus indirect neurogenesis. These findings have Moulin INSERM U1270, Paris, implications for understanding cortical malformations in ciliopathies with INPP5E mutations.
    [Show full text]
  • The Dual Roles of Betacellulin and the ERBB Receptors in Acute Pancreatitis & Pancreatic Ductal Adenocarcinoma – a Mouse Study
    Dissertation zur Erlangung des Doktorgrades der Fakultät für Chemie und Pharmazie der Ludwig-Maximilians-Universität München The dual roles of betacellulin and the ERBB receptors in acute pancreatitis & pancreatic ductal adenocarcinoma – a mouse study Kathrin Verena Hedegger aus Frankfurt am Main 2019 Erklärung Diese Dissertation wurde im Sinne von § 7 der Promotionsordnung vom 28. November 2011 von Herrn Prof. Dr. Eckhard Wolf betreut und von Herrn PD Dr. Dietmar Martin von der Fakultät für Chemie und Pharmazie vertreten. Eidesstattliche Versicherung Diese Dissertation wurde eigenständig und ohne unerlaubte Hilfe erarbeitet. München, 02.08.2019, Kathrin Verena Hedegger __________________________________ Dissertation eingereicht am: 02.08.2019 1. Gutachter: PD Dr. Dietmar Martin 2. Gutachter: Prof. Dr. Eckhard Wolf Mündliche Prüfung am: 13.09.2019 THE DUAL ROLES OF BETACELLULIN AND THE ERBB RECEPTORS IN ACUTE PANCREATITIS & PANCREATIC DUCTAL ADENOCARCINOMA – A MOUSE STUDY DANKSAGUNG Ein kleines Dankeschön an alle, die sowohl direkt, als auch indirekt an dieser Arbeit mitgewirkt haben. Mein Dank geht zunächst an meinen Doktorvater PD. Dr. Dietmar Martin für die Bereitschaft meine Dissertation an der Fakultät Chemie und Pharmazie zu betreuen, für die gute Zusammenarbeit und seine Ratschläge. Herrn Prof. Dr. Eckhard Wolf möchte ich für die Übernahme des Zweitgutachtens danken, sowie für die Bereitstellung seines Labors und für die zahlreichen Diskussionen und Anregungen. Mein Dank gilt außerdem Herrn PD Dr. Dietmar Martin, Herrn Prof. Dr. Eckhard Wolf, Herrn PD Dr. Maik Dahlhoff, Herrn Prof. Dr. Klaus Förstemann, Herrn Prof. Dr. Julian Stingele und Herrn Prof. Dr. Karl-Peter Hopfner für die Bereitschaft mein Prüfungskomitee zu bilden. PD Dr. Maik Dahlhoff möchte ich ganz herzlich für die Betreuung meiner Doktorarbeit danken, zunächst für die Auswahl der Projekte, vor allem aber für seine Unterstützung über all die Jahre.
    [Show full text]
  • ARL13B, PDE6D, and CEP164 Form a Functional Network for INPP5E Ciliary Targeting
    ARL13B, PDE6D, and CEP164 form a functional network for INPP5E ciliary targeting Melissa C. Humberta,b, Katie Weihbrechta,b, Charles C. Searbyb,c, Yalan Lid, Robert M. Poped, Val C. Sheffieldb,c, and Seongjin Seoa,1 aDepartment of Ophthalmology and Visual Sciences, bDepartment of Pediatrics, cHoward Hughes Medical Institute, and dProteomics Facility, University of Iowa, Iowa City, IA 52242 Edited by Kathryn V. Anderson, Sloan-Kettering Institute, New York, NY, and approved October 19, 2012 (received for review June 28, 2012) Mutations affecting ciliary components cause a series of related polydactyly, skeletal defects, cleft palate, and cerebral develop- genetic disorders in humans, including nephronophthisis (NPHP), mental defects (11). Inactivation of Inpp5e in adult mice results in Joubert syndrome (JBTS), Meckel-Gruber syndrome (MKS), and Bar- obesity and photoreceptor degeneration. Interestingly, many pro- det-Biedl syndrome (BBS), which are collectively termed “ciliopa- teins that localize to cilia, including INPP5E, RPGR, PDE6 α and thies.” Recent protein–protein interaction studies combined with β subunits, GRK1 (Rhodopsin kinase), and GNGT1 (Transducin γ genetic analyses revealed that ciliopathy-related proteins form sev- chain), are prenylated (either farnesylated or geranylgeranylated), eral functional networks/modules that build and maintain the pri- and mutations in these genes or genes involved in their prenylation mary cilium. However, the precise function of many ciliopathy- (e.g., AIPL1 and RCE1) lead to photoreceptor
    [Show full text]
  • Downloaded October, 2015) 83
    bioRxiv preprint doi: https://doi.org/10.1101/522607; this version posted January 17, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Induction of an alternative 5’ leader enhances translation of Inpp5e and resistance to oncolytic virus infection Huy-Dung Hoang1,Δ, Tyson E. Graber1,2,Δ, Jian-Jun Jia1, Nasana Vaidya1, Victoria Gilchrist1, Wencheng Li3, Christos G. Gkogkas4, Maritza Jaramillo5, Seyed Mehdi Jafarnejad6, and Tommy Alain1* 1 Children's Hospital of Eastern Ontario Research Institute, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada 2 Department of Biochemistry and Goodman Cancer Center, McGill University, Montreal, Quebec, Canada 3 Department of Biochemistry and Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, USA 4 Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK. 5 INRS Institut Armand-Frappier Research Centre, Laval, Quebec, Canada 6 Centre for Cancer Research and Cell Biology, The Queen’s University of Belfast, Belfast, UK *correspondence: Tommy Alain Children’s Hospital of Eastern Ontario Research Institute 401 Smyth Road Ottawa, Ontario, Canada Δ These authors contributed equally to this work. Short Title: Oncolytic viruses trigger an innate RNA translation response ABSTRACT variant that lacks repressive upstream open reading frames (uORFs) within its 5’ leader and is consequently Residual cell-intrinsic innate immunity in cancer cells efficiently translated. Furthermore, we show that INPP5E hampers infection with oncolytic viruses. mRNA contributes to antiviral immunity by altering virus translation is an important component of innate attachment. These findings uncover a role for immunity, yet the targeted cellular mRNAs remain ill- translational control through alternative 5’ leader defined.
    [Show full text]
  • Live-Cell Imaging Rnai Screen Identifies PP2A–B55α and Importin-Β1 As Key Mitotic Exit Regulators in Human Cells
    LETTERS Live-cell imaging RNAi screen identifies PP2A–B55α and importin-β1 as key mitotic exit regulators in human cells Michael H. A. Schmitz1,2,3, Michael Held1,2, Veerle Janssens4, James R. A. Hutchins5, Otto Hudecz6, Elitsa Ivanova4, Jozef Goris4, Laura Trinkle-Mulcahy7, Angus I. Lamond8, Ina Poser9, Anthony A. Hyman9, Karl Mechtler5,6, Jan-Michael Peters5 and Daniel W. Gerlich1,2,10 When vertebrate cells exit mitosis various cellular structures can contribute to Cdk1 substrate dephosphorylation during vertebrate are re-organized to build functional interphase cells1. This mitotic exit, whereas Ca2+-triggered mitotic exit in cytostatic-factor- depends on Cdk1 (cyclin dependent kinase 1) inactivation arrested egg extracts depends on calcineurin12,13. Early genetic studies in and subsequent dephosphorylation of its substrates2–4. Drosophila melanogaster 14,15 and Aspergillus nidulans16 reported defects Members of the protein phosphatase 1 and 2A (PP1 and in late mitosis of PP1 and PP2A mutants. However, the assays used in PP2A) families can dephosphorylate Cdk1 substrates in these studies were not specific for mitotic exit because they scored pro- biochemical extracts during mitotic exit5,6, but how this relates metaphase arrest or anaphase chromosome bridges, which can result to postmitotic reassembly of interphase structures in intact from defects in early mitosis. cells is not known. Here, we use a live-cell imaging assay and Intracellular targeting of Ser/Thr phosphatase complexes to specific RNAi knockdown to screen a genome-wide library of protein substrates is mediated by a diverse range of regulatory and targeting phosphatases for mitotic exit functions in human cells. We subunits that associate with a small group of catalytic subunits3,4,17.
    [Show full text]
  • Phosphatases Page 1
    Phosphatases esiRNA ID Gene Name Gene Description Ensembl ID HU-05948-1 ACP1 acid phosphatase 1, soluble ENSG00000143727 HU-01870-1 ACP2 acid phosphatase 2, lysosomal ENSG00000134575 HU-05292-1 ACP5 acid phosphatase 5, tartrate resistant ENSG00000102575 HU-02655-1 ACP6 acid phosphatase 6, lysophosphatidic ENSG00000162836 HU-13465-1 ACPL2 acid phosphatase-like 2 ENSG00000155893 HU-06716-1 ACPP acid phosphatase, prostate ENSG00000014257 HU-15218-1 ACPT acid phosphatase, testicular ENSG00000142513 HU-09496-1 ACYP1 acylphosphatase 1, erythrocyte (common) type ENSG00000119640 HU-04746-1 ALPL alkaline phosphatase, liver ENSG00000162551 HU-14729-1 ALPP alkaline phosphatase, placental ENSG00000163283 HU-14729-1 ALPP alkaline phosphatase, placental ENSG00000163283 HU-14729-1 ALPPL2 alkaline phosphatase, placental-like 2 ENSG00000163286 HU-07767-1 BPGM 2,3-bisphosphoglycerate mutase ENSG00000172331 HU-06476-1 BPNT1 3'(2'), 5'-bisphosphate nucleotidase 1 ENSG00000162813 HU-09086-1 CANT1 calcium activated nucleotidase 1 ENSG00000171302 HU-03115-1 CCDC155 coiled-coil domain containing 155 ENSG00000161609 HU-09022-1 CDC14A CDC14 cell division cycle 14 homolog A (S. cerevisiae) ENSG00000079335 HU-11533-1 CDC14B CDC14 cell division cycle 14 homolog B (S. cerevisiae) ENSG00000081377 HU-06323-1 CDC25A cell division cycle 25 homolog A (S. pombe) ENSG00000164045 HU-07288-1 CDC25B cell division cycle 25 homolog B (S. pombe) ENSG00000101224 HU-06033-1 CDKN3 cyclin-dependent kinase inhibitor 3 ENSG00000100526 HU-02274-1 CTDSP1 CTD (carboxy-terminal domain,
    [Show full text]
  • Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss
    cells Review Mouse Models of Inherited Retinal Degeneration with Photoreceptor Cell Loss 1, 1, 1 1,2,3 1 Gayle B. Collin y, Navdeep Gogna y, Bo Chang , Nattaya Damkham , Jai Pinkney , Lillian F. Hyde 1, Lisa Stone 1 , Jürgen K. Naggert 1 , Patsy M. Nishina 1,* and Mark P. Krebs 1,* 1 The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA; [email protected] (G.B.C.); [email protected] (N.G.); [email protected] (B.C.); [email protected] (N.D.); [email protected] (J.P.); [email protected] (L.F.H.); [email protected] (L.S.); [email protected] (J.K.N.) 2 Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand 3 Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand * Correspondence: [email protected] (P.M.N.); [email protected] (M.P.K.); Tel.: +1-207-2886-383 (P.M.N.); +1-207-2886-000 (M.P.K.) These authors contributed equally to this work. y Received: 29 February 2020; Accepted: 7 April 2020; Published: 10 April 2020 Abstract: Inherited retinal degeneration (RD) leads to the impairment or loss of vision in millions of individuals worldwide, most frequently due to the loss of photoreceptor (PR) cells. Animal models, particularly the laboratory mouse, have been used to understand the pathogenic mechanisms that underlie PR cell loss and to explore therapies that may prevent, delay, or reverse RD. Here, we reviewed entries in the Mouse Genome Informatics and PubMed databases to compile a comprehensive list of monogenic mouse models in which PR cell loss is demonstrated.
    [Show full text]