Reprogramming the Retina: Next Generation Strategies of Retinal Neuroprotection and Gene Therapy Vector Potency Assessment

Total Page:16

File Type:pdf, Size:1020Kb

Reprogramming the Retina: Next Generation Strategies of Retinal Neuroprotection and Gene Therapy Vector Potency Assessment University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2018 Reprogramming The Retina: Next Generation Strategies Of Retinal Neuroprotection And Gene Therapy Vector Potency Assessment Devin Scott Mcdougald University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Genetics Commons, Molecular Biology Commons, and the Virology Commons Recommended Citation Mcdougald, Devin Scott, "Reprogramming The Retina: Next Generation Strategies Of Retinal Neuroprotection And Gene Therapy Vector Potency Assessment" (2018). Publicly Accessible Penn Dissertations. 3158. https://repository.upenn.edu/edissertations/3158 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/3158 For more information, please contact [email protected]. Reprogramming The Retina: Next Generation Strategies Of Retinal Neuroprotection And Gene Therapy Vector Potency Assessment Abstract Mutations within over 250 known genes are associated with inherited retinal degeneration. Clinical success following gene replacement therapy for Leber’s congenital amaurosis type 2 establishes a platform for the development of downstream treatments targeting other forms of inherited and acquired ocular disease. Unfortunately, several challenges relevant to complex disease pathology and limitations of current gene transfer technologies impede the development of gene replacement for each specific form of retinal degeneration. Here we describe gene augmentation strategies mediated by recombinant AAV vectors that impede retinal degeneration in pre-clinical models of acquired and inherited vision loss. We demonstrate distinct neuroprotective effects upon retinal ganglion cell survival and function in experimental optic neuritis following AAV-mediated gene augmentation. Gene transfer of the antioxidant transcription factor, NRF2, improves RGC survival while overexpression of the pro-survival and anti- inflammatory protein, SIRT1, promotes preservation of visual function. In the context of inherited retinal disease, we show stimulation of anabolic metabolism following AKT3 gene transfer preserves photoreceptor viability and delays functional loss in a mouse model of retinitis pigmentosa. In addition to these neuroprotective strategies, we also describe an approach to improve the in vitro potency of AAV vectors that are restricted by tissue-specific regulatory elements. This strategy utilizes genome engineering based on CRISPR/Cas9 gene activation to reprogram cell lines to specifically and potently express tissue-specific promoters of interest from AAV vectors. Degree Type Dissertation Degree Name Doctor of Philosophy (PhD) Graduate Group Cell & Molecular Biology First Advisor Jean Bennett Keywords aav, gene therapy, neuroprotection, photoreceptor, retinal degeneration, retinal ganglion cell Subject Categories Genetics | Molecular Biology | Virology This dissertation is available at ScholarlyCommons: https://repository.upenn.edu/edissertations/3158 REPROGRAMMING THE RETINA: NEXT GENERATION STRAEGIES OF RETINAL NEUROPROTECTION AND GENE THERAPY VECTOR POTENCY ASSESSMENT Devin S. McDougald A DISSERTATION in Cell & Molecular Biology Presented to the Faculties of the University of Pennsylvania in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy 2018 Supervisor of Dissertation ____________________ Jean Bennett M.D., Ph.D., Professor of Ophthalmology and Cell & Developmental Biology Graduate Group Chairperson _____________________ Dan Kessler Ph.D., Chair, Cell & Molecular Biology Graduate Group Dissertation Committee Valder Arruda M.D., Ph.D. Claire H. Mitchell Associate Professor of Pediatrics Professor of Anatomy & Cell Biology Kenneth S. Shindler M.D., Ph.D. Hansell H. Stedman Associate Professor of Ophthalmology Associate Professor of Surgery i ACKNOWLEDGMENT I would have not reached this point academically, professionally, or personally without the influence of past scientific mentors. Dr. Anthony Bell Jr. at the University of Southern Mississippi presented my first opportunity to perform laboratory research. His guidance as well as patience provided an excellent foundation in molecular biology techniques, experimental design, and how to conduct oneself within a research lab. Drs. Wade Chien, Lisa Cunningham, and Thomas Friedman at the NIDCD/NIH allowed me to train in an exceptionally fast-paced and cutting edge research environment prior to entering graduate school. Observing their passion, dedication, and relentless work ethic towards improving both the fundamental understanding of human diseases and creation of novel therapeutics reinvigorated my passion for pursuit of a research-based career. I have been incredibly fortunate to carry out this work alongside a crew of exceptionally talented scientists from very diverse training backgrounds. I want to particularly acknowledge Drs. Adam Wojno, Scott Dooley, Kris Fisher, Jason Mills, and Pavitra Ramachandran for always lending their time for insightful discussions or venting sessions relevant to project design, troubleshooting, career advice, and life. Much of the work shown here could not have been performed without the technical expertise of many individuals including Zhangyong Wei, Alexandra Zezulin, Kimberly Dine, Tyler Papp, and Dr. Ken Shindler. In addition, I must thank members of the CAROT research vector core, especially Drs. Shangzhen Zhou and Jieyan Pan, for producing the AAV vectors used in these studies. I thank my thesis committee for their helpful insight and technical direction in guiding some of the studies presented in this document. The ideas and concepts behind much of this work would likely have been shot down immediately if I were under the direction of any other scientist besides Jean Bennett. I believe this is one of the many reasons Jean has sustained a highly productive scientific career: she places an incredible amount of trust in her trainees to independently develop and explore creative ideas and solutions to challenging problems in vision research. Beyond all the scientific achievements and medical milestones scattered along her career path, Jean maintains an unwavering humble, ii supportive, and congenial nature of a magnitude that I have yet to encounter elsewhere in the research community. Altogether, she is an exceptional scientist and an even better person, and I am forever grateful to her for allowing me to train and grow as a scientist in her laboratory. I must thank my family and friends for their enduring love and support throughout this very long and tiresome process. I want to specifically acknowledge Terra Kuhn, Bobby French, Steven Zhao, Stephen Bart, Chris Cali, David Walter, and Lucas Van Gorder for their friendship and overwhelming support during the best and worst of times. Finally, I must thank my parents, Glenn and Elizabeth, for their continued support through all avenues of life. iii ABSTRACT REPROGRAMMING THE RETINA: NEXT GENERATION STRATEGIES OF RETINAL NEUROPROTECTION AND GENE THERAPY VECTOR POTENCY ASSESSMENT Devin S. McDougald Jean Bennett M.D., Ph.D. Mutations within over 250 known genes are associated with inherited retinal degeneration. Clinical success following gene replacement therapy for Leber’s congenital amaurosis type 2 establishes a platform for the development of downstream treatments targeting other forms of inherited and acquired ocular disease. Unfortunately, several challenges relevant to complex disease pathology and limitations of current gene transfer technologies impede the development of gene replacement for each specific form of retinal degeneration. Here we describe gene augmentation strategies mediated by recombinant AAV vectors that impede retinal degeneration in pre-clinical models of acquired and inherited vision loss. We demonstrate distinct neuroprotective effects upon retinal ganglion cell survival and function in experimental optic neuritis following AAV-mediated gene augmentation. Gene transfer of the antioxidant transcription factor, NRF2, improves RGC survival while overexpression of the pro-survival and anti-inflammatory protein, SIRT1, promotes preservation of visual function. In the context of inherited retinal disease, we show stimulation of anabolic metabolism following AKT3 gene transfer preserves photoreceptor viability and delays functional loss in a mouse model of retinitis pigmentosa. In addition to these neuroprotective strategies, we also describe an approach to improve the in vitro potency of AAV vectors that are restricted by tissue-specific regulatory elements. This strategy utilizes genome engineering based on CRISPR/Cas9 gene activation to reprogram cell lines to specifically and potently express tissue-specific promoters of interest from AAV vectors. iv TABLE OF CONTENTS ACKNOWLEDGMENT ............................................................................................................. I ABSTRACT ............................................................................................................................... IV LIST OF FIGURES .................................................................................................................. VI CHAPTER 1 INTRODUCTION ............................................................................................. 1 CHAPTER 2 STIMULATING ANTI-INFLAMMATORY/ANTIOXIDANT PATHWAYS IN EXPERIMENTAL OPTIC NEURITIS WITH AAV-MEDIATED GENE TRANSFER ................................................................................................................
Recommended publications
  • Table 2. Significant
    Table 2. Significant (Q < 0.05 and |d | > 0.5) transcripts from the meta-analysis Gene Chr Mb Gene Name Affy ProbeSet cDNA_IDs d HAP/LAP d HAP/LAP d d IS Average d Ztest P values Q-value Symbol ID (study #5) 1 2 STS B2m 2 122 beta-2 microglobulin 1452428_a_at AI848245 1.75334941 4 3.2 4 3.2316485 1.07398E-09 5.69E-08 Man2b1 8 84.4 mannosidase 2, alpha B1 1416340_a_at H4049B01 3.75722111 3.87309653 2.1 1.6 2.84852656 5.32443E-07 1.58E-05 1110032A03Rik 9 50.9 RIKEN cDNA 1110032A03 gene 1417211_a_at H4035E05 4 1.66015788 4 1.7 2.82772795 2.94266E-05 0.000527 NA 9 48.5 --- 1456111_at 3.43701477 1.85785922 4 2 2.8237185 9.97969E-08 3.48E-06 Scn4b 9 45.3 Sodium channel, type IV, beta 1434008_at AI844796 3.79536664 1.63774235 3.3 2.3 2.75319499 1.48057E-08 6.21E-07 polypeptide Gadd45gip1 8 84.1 RIKEN cDNA 2310040G17 gene 1417619_at 4 3.38875643 1.4 2 2.69163229 8.84279E-06 0.0001904 BC056474 15 12.1 Mus musculus cDNA clone 1424117_at H3030A06 3.95752801 2.42838452 1.9 2.2 2.62132809 1.3344E-08 5.66E-07 MGC:67360 IMAGE:6823629, complete cds NA 4 153 guanine nucleotide binding protein, 1454696_at -3.46081884 -4 -1.3 -1.6 -2.6026947 8.58458E-05 0.0012617 beta 1 Gnb1 4 153 guanine nucleotide binding protein, 1417432_a_at H3094D02 -3.13334396 -4 -1.6 -1.7 -2.5946297 1.04542E-05 0.0002202 beta 1 Gadd45gip1 8 84.1 RAD23a homolog (S.
    [Show full text]
  • Protein Identities in Evs Isolated from U87-MG GBM Cells As Determined by NG LC-MS/MS
    Protein identities in EVs isolated from U87-MG GBM cells as determined by NG LC-MS/MS. No. Accession Description Σ Coverage Σ# Proteins Σ# Unique Peptides Σ# Peptides Σ# PSMs # AAs MW [kDa] calc. pI 1 A8MS94 Putative golgin subfamily A member 2-like protein 5 OS=Homo sapiens PE=5 SV=2 - [GG2L5_HUMAN] 100 1 1 7 88 110 12,03704523 5,681152344 2 P60660 Myosin light polypeptide 6 OS=Homo sapiens GN=MYL6 PE=1 SV=2 - [MYL6_HUMAN] 100 3 5 17 173 151 16,91913397 4,652832031 3 Q6ZYL4 General transcription factor IIH subunit 5 OS=Homo sapiens GN=GTF2H5 PE=1 SV=1 - [TF2H5_HUMAN] 98,59 1 1 4 13 71 8,048185945 4,652832031 4 P60709 Actin, cytoplasmic 1 OS=Homo sapiens GN=ACTB PE=1 SV=1 - [ACTB_HUMAN] 97,6 5 5 35 917 375 41,70973209 5,478027344 5 P13489 Ribonuclease inhibitor OS=Homo sapiens GN=RNH1 PE=1 SV=2 - [RINI_HUMAN] 96,75 1 12 37 173 461 49,94108966 4,817871094 6 P09382 Galectin-1 OS=Homo sapiens GN=LGALS1 PE=1 SV=2 - [LEG1_HUMAN] 96,3 1 7 14 283 135 14,70620005 5,503417969 7 P60174 Triosephosphate isomerase OS=Homo sapiens GN=TPI1 PE=1 SV=3 - [TPIS_HUMAN] 95,1 3 16 25 375 286 30,77169764 5,922363281 8 P04406 Glyceraldehyde-3-phosphate dehydrogenase OS=Homo sapiens GN=GAPDH PE=1 SV=3 - [G3P_HUMAN] 94,63 2 13 31 509 335 36,03039959 8,455566406 9 Q15185 Prostaglandin E synthase 3 OS=Homo sapiens GN=PTGES3 PE=1 SV=1 - [TEBP_HUMAN] 93,13 1 5 12 74 160 18,68541938 4,538574219 10 P09417 Dihydropteridine reductase OS=Homo sapiens GN=QDPR PE=1 SV=2 - [DHPR_HUMAN] 93,03 1 1 17 69 244 25,77302971 7,371582031 11 P01911 HLA class II histocompatibility antigen,
    [Show full text]
  • Renewed Momentum in Ocular Gene and Cell Therapy, Broadening Application to Chronic Diseases
    FEATURE Renewed momentum in ocular gene and cell therapy, broadening application to chronic diseases BY ROD MCNEIL Gene and cell therapies offer the prospect of ground-breaking new avenues for the treatment of diseases, reflected in a renewed explosion of interest and investment in retinal gene therapy. Rod McNeil reports recent clinical trial readouts across a diverse range of investigational ocular gene and cell therapy candidates. ene therapy is literally giving transfer clinical trials to date involving (VA) at 24 months in patients treated with sight to children who would subretinal and intravitreal delivery. The timrepigene emparvovec compared with otherwise not see,” said Dr majority of these studies use an adeno- untreated patients in the natural history GJean Bennett, delivering the associated virus (AAV) vector. study. At two years over 90% of patients “ treated with timrepigene emparvovec Charles L Schepens MD Lecture jointly with Prof Albert Maguire at the American Gene therapy for choroideremia maintained VA. In a subset of treated Academy of Ophthalmology 2019 Retina Investigational gene therapy timrepigene patients with moderate to severe VA loss, Subspecialty Day. Dr Bennett has developed emparvovec (BIIB111/AAV2-REP1, Biogen) 21% experienced a VA improvement of at gene transfer approaches to test treatment is an AAV2 vector administered by least 15 letters from baseline compared with strategies for retinal degenerative and subretinal injection being evaluated as a 1.0% of untreated patients. ocular neovascular diseases and her work treatment for choroideremia (CHM). Biogen led to the first approved gene therapy announced November 2019 completion GenSight Biologics targets novel product targeting a retinal disease of patient enrolment in the global phase gene therapies for LHON and worldwide.” 3 STAR clinical trial of 170 adult males retinitis pigmentosa patients Gene therapy has definitely arrived.
    [Show full text]
  • Stem Cells Set Their Sights on Retinitis Pigmentosa
    INSIGHT elife.elifesciences.org OPHTHALMOLOGY Stem cells set their sights on retinitis pigmentosa Skin cells from a patient with a form of inherited blindness have been reprogrammed into retinal cells and successfully transplanted into mice. JEANNETTE L BENNICELLI AND JEAN BENNETT loss to identify the genetic mutations leading Related research article Tucker BA, to their blindness; the Iowa team also generate induced pluripotent stem cells (iPSCs) from these Mullins RF, Streb LM, Anfinson K, Eyestone individuals to create patient-specific models of ME, Kaalberg E, Riker MJ, Drack AV, Braun disease. Now, in eLife, Stone and co-workers— TA, Stone EM. 2013. Patient-specific including Budd Tucker as first author—report that iPSC-derived photoreceptor precursor they have used stem cell technology to create a personalized model of a recessive form of retinitis cells as a means to investigate retinitis pigmentosa, and that they have also successfully pigmentosa. eLife 2:e00824. doi: 10.7554/ transplanted the cells into mice (Tucker et al., eLife.00824 2013). These results are an important step toward Image Photoreceptors derived from human autologous transplantation, the regeneration of tissues damaged by disease using stem cells stem cells can colonize a mouse retina derived from the patient’s own cells (Figure 1). (arrow) In addition to benefiting basic research, these findings represent a means to develop specific understanding of, and treatment for, a range of genetic conditions—in particular, the large set of nherited blindness encompasses a wide highly idiosyncratic syndromes that constitute spectrum of pathologies that can be caused inherited blindness. Iby mutations in more than 220 genes.
    [Show full text]
  • Rhodopsin Kinase Activity Modulates the Amplitude of the Visual Response in Drosophila
    Rhodopsin kinase activity modulates the amplitude of the visual response in Drosophila Seung-Jae Lee*, Hong Xu, and Craig Montell† Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205 Edited by Robert J. Lefkowitz, Duke University Medical Center, Durham, NC, and approved June 30, 2004 (received for review March 29, 2004) A feature shared between Drosophila rhodopsin and nearly all tractability of fly genetics (14). As in mammals, light-activated other G protein-coupled receptors is agonist-dependent protein rhodopsin is phosphorylated and interacts with a protein, arres- phosphorylation. Despite extensive analyses of Drosophila photo- tin, which facilitates deactivation of the receptor. However, transduction, the identity and function of the rhodopsin kinase unlike mammalian phototransduction, light activation in Dro- (RK) have been elusive. Here, we provide evidence that G protein- sophila is coupled to stimulation of phospholipase C rather than coupled receptor kinase 1 (GPRK1), which is most similar to the a cGMP-phosphodiesterase. ␤-adrenergic receptor kinases, G protein-coupled receptor kinase 2 Two Drosophila genes encoding putative GRKs (GPRK1 and (GRK2) and GRK3, is the fly RK. We show that GPRK1 is enriched in GPRK2) were isolated more than a decade ago (15), but were photoreceptor cells, associates with the major Drosophila rhodop- dismissed as candidate RKs due in part to their much greater sin, Rh1, and phosphorylates the receptor. As is the case with similarity to mammalian nonvisual GRKs than to mammalian mammalian GRK2 and GRK3, Drosophila GPRK1 includes a C- RKs. Subsequent analysis of GPRK2 demonstrated that it is terminal pleckstrin homology domain, which binds to phosphoi- expressed in the ovaries and required for egg morphogenesis nositides and the G␤␥ subunit.
    [Show full text]
  • Balancing the Photoreceptor Proteome: Proteostasis Network Therapeutics for Inherited Retinal Disease
    G C A T T A C G G C A T genes Review Balancing the Photoreceptor Proteome: Proteostasis Network Therapeutics for Inherited Retinal Disease Siebren Faber and Ronald Roepman * Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands * Correspondence: [email protected] Received: 10 June 2019; Accepted: 16 July 2019; Published: 24 July 2019 Abstract: The light sensing outer segments of photoreceptors (PRs) are renewed every ten days due to their high photoactivity, especially of the cones during daytime vision. This demands a tremendous amount of energy, as well as a high turnover of their main biosynthetic compounds, membranes, and proteins. Therefore, a refined proteostasis network (PN), regulating the protein balance, is crucial for PR viability. In many inherited retinal diseases (IRDs) this balance is disrupted leading to protein accumulation in the inner segment and eventually the death of PRs. Various studies have been focusing on therapeutically targeting the different branches of the PR PN to restore the protein balance and ultimately to treat inherited blindness. This review first describes the different branches of the PN in detail. Subsequently, insights are provided on how therapeutic compounds directed against the different PN branches might slow down or even arrest the appalling, progressive blinding conditions. These insights are supported by findings of PN modulators in other research disciplines. Keywords: protein trafficking; protein folding; protein degradation; chaperones; chaperonins; heat shock response; unfolded protein response; autophagy; therapy 1. Introduction The rod and cone photoreceptor (PR) cells are the most abundant cell types in the human retina, with ~6.4 million cones and up to 125 million rods per adult retina [1].
    [Show full text]
  • Scheie Vision Department of Opthalmology
    summer 2018 scheie vision Department of Opthalmology Like Watching a Miracle: From Landmark Gene Therapy to the Stage of America’s Got Talent IN THIS ISSUE A MESSAGE FROM THE CHAIR Dear Friends, VISION Penn Medicine’s Department of Ophthalmology, Scheie Eye Institute, is dedicated to cutting edge research, 02 Like Watching a Miracle providing the highest quality of care in Philadelphia and around the world, and training the next generation 04 Landmark FDA Approval of ophthalmologists. Our faculty and staff strive to cultivate an environment of continued learning and 08 Studying Individual Photoreceptors mentoring, where young minds with great potential grow and thrive. Our alumni go on to lead impactful 10 Intraocular Bleeding from careers, maintaining relationships with peers and mentors and returning to the Annual Alumni Meeting Blood Clot Meds? each spring. This event is always a reminder of the outstanding accomplishments of Scheie’s alumni, 11 New Options for Dry Eye students, staff, and faculty, and their daily commitment to improving the lives of patients and colleagues. This issue of Scheie Vision covers the people behind SCHEIE COMMUNITY Scheie’s advances and mission of excellence. We 13 Beautiful Inside and Out feature Lang Lourng Ung, an ophthalmic technician who brings inspirational resilience and passion to working with patients; Sonul Mehta, MD, who travels 15 Faces of Scheie around the world to provide ophthalmic care in underserved communities; Jessica Morgan, PhD, whose 19 Eye Care Across the World research on photoreceptor function has tremendous implications for the diagnosis and treatment of retinal 20 Remembering Walker Kirby disease; and Jean Bennett, MD, PhD, and Al Maguire, MD, who have demonstrated unwavering commitment for over 25 years to making it possible for blind 21 144th Anniversary Weekend children to see.
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2010/0158968 A1 Panitch Et Al
    US 20100158968A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2010/0158968 A1 Panitch et al. (43) Pub. Date: Jun. 24, 2010 (54) CELL-PERMEANT PEPTIDE-BASED Publication Classification INHIBITOR OF KINASES (51) Int. Cl. (76) Inventors: Alyssa Panitch, West Lafayette, IN st e8 CR (US); Brandon Seal, West ( .01) Lafayette, IN (US) A638/10 (2006.01) s A638/16 (2006.01) Correspondence Address: A6IP 43/00 (2006.01) GREENBERG TRAURIG, LLP (52) U.S. Cl. ................ 424/422:514/15: 514/13: 514/14 200 PARKAVE., P.O. BOX 677 FLORHAMPARK, NJ 07932 (US) (57) ABSTRACT The described invention provides kinase inhibiting composi (21) Appl. No.: 12/634,476 tions containing a therapeutic amount of a therapeutic inhibi (22) Filed: Dec. 9, 2009 torpeptide that inhibits at least one kinase enzyme, methods e 19 for treating an inflammatory disorder whose pathophysiology comprises inflammatory cytokine expression, and methods Related U.S. Application Data for treating an inflammatory disorder whose pathophysiology (60) Provisional application No. 61/121,396, filed on Dec. comprises inflammatory cytokine expression using the kinase 10, 2008. inhibiting compositions. 20 { ki> | 0: & c s - --- 33- x: SE PEPELE, ics 1.-- E- X K. AAA 22.9 --- KKK. Y.A., 3.2; C. -r { AAEASA. A. E. i : A X AAAAAAA; ; ; ; :-n. 4:-: is SEEKESAN.ARESA, 3523 -- -- Yili.A.R.AKA: 5,342 3. {{RCE: Rix i: Patent Application Publication US 2010/0158968A1 & ******** NO s ***** · Patent Application Publication Jun. 24, 2010 Sheet 2 of 11 US 2010/0158968A1 it, O Peptide: Cso: g E 100 WRRKAWRRKANRO, GWAA.
    [Show full text]
  • This Thesis Has Been Submitted in Fulfilment of the Requirements for a Postgraduate Degree (E.G
    This thesis has been submitted in fulfilment of the requirements for a postgraduate degree (e.g. PhD, MPhil, DClinPsychol) at the University of Edinburgh. Please note the following terms and conditions of use: This work is protected by copyright and other intellectual property rights, which are retained by the thesis author, unless otherwise stated. A copy can be downloaded for personal non-commercial research or study, without prior permission or charge. This thesis cannot be reproduced or quoted extensively from without first obtaining permission in writing from the author. The content must not be changed in any way or sold commercially in any format or medium without the formal permission of the author. When referring to this work, full bibliographic details including the author, title, awarding institution and date of the thesis must be given. Molecular mechanisms underlying Retinitis pigmentosa type 2 Rodanthi Lyraki Thesis submitted for the degree of Doctor of Philosophy University of Edinburgh 2017 Declaration I declare that this thesis is my own work, and that the experiments described here were conducted by me except where explicitly stated. This work has not been submitted for any other degree or professional qualification. Rodanthi Lyraki, August 2017 ii Preface “Photoreceptors sit on a knife edge separating function and survival from dysfunction and death, and almost any defect seems capable of tipping them towards cell death.” - Alan F. Wright et al., “Photoreceptor degeneration: genetic and mechanistic dissection of a complex trait” iii Acknowledgements I feel very fortunate to have carried out my PhD in the Institute of Genetics and Molecular Medicine in Edinburgh, where I had the opportunity to interact with first- class scientists on a daily basis.
    [Show full text]
  • Novel Adeno-Associated Viral Vectors for Retinal Gene Therapy
    Gene Therapy (2012) 19, 162–168 & 2012 Macmillan Publishers Limited All rights reserved 0969-7128/12 www.nature.com/gt REVIEW Novel adeno-associated viral vectors for retinal gene therapy This article has been corrected since Advance Online Publication and an erratum is also printed in this issue LH Vandenberghe1 and A Auricchio2,3 Vectors derived from adeno-associated virus (AAV) are currently the most promising vehicles for therapeutic gene delivery to the retina. Recently, subretinal administration of AAV2 has been demonstrated to be safe and effective in patients with a rare form of inherited childhood blindness, suggesting that AAV-mediated retinal gene therapy may be successfully extended to other blinding conditions. This is further supported by the great versatility of AAV as a vector platform as there are a large number of AAV variants and many of these have unique transduction characteristics useful for targeting different cell types in the retina including glia, epithelium and many types of neurons. Naturally occurring, rationally designed or in vitro evolved AAV vectors are currently being utilized to transduce several different cell types in the retina and to treat a variety of animal models of retinal disease. The continuous and creative development of AAV vectors provides opportunities to overcome existing challenges in retinal gene therapy such as efficient transfer of genes exceeding AAV’s cargo capacity, or the targeting of specific cells within the retina or transduction of photoreceptors following routinely used intravitreal
    [Show full text]
  • Abnormal Photoresponses and Light-Induced Apoptosis in Rods Lacking Rhodopsin Kinase
    Proc. Natl. Acad. Sci. USA Vol. 96, pp. 3718–3722, March 1999 Cell Biology Abnormal photoresponses and light-induced apoptosis in rods lacking rhodopsin kinase CHING-KANG CHEN*†,MARIE E. BURNS†‡,MARIBETH SPENCER§,GREGORY A. NIEMI§,JEANNIE CHEN¶, i JAMES B. HURLEY§,DENIS A. BAYLOR‡, AND MELVIN I. SIMON* *Division of Biology, 147-75, California Institute of Technology, Pasadena, CA 91125; ‡Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305; §Howard Hughes Medical Institute and Department of Biochemistry, Box 357370, University of Washington, Seattle, WA 98195; and ¶Department of Cell and Neurobiology, University of Southern California, Los Angeles, CA 90033 Contributed by Melvin Simon, January 12, 1999 ABSTRACT Phosphorylation is thought to be an essential 10) and phosphorylate rhodopsin’s C-terminal residues equally first step in the prompt deactivation of photoexcited rhodop- well in vitro (11), it is unclear which kinase is mainly responsible sin. In vitro, the phosphorylation can be catalyzed either by for rhodopsin deactivation in vivo. We determined the role of rhodopsin kinase (RK) or by protein kinase C (PKC). To RK in rhodopsin deactivation in intact rods by deactivating investigate the specific role of RK, we inactivated both alleles both alleles of the RK gene. We found that RK is required for of the RK gene in mice. This eliminated the light-dependent the normal deactivation of rhodopsin and that in its absence, phosphorylation of rhodopsin and caused the single-photon dramatic functional and structural changes occurred. response to become larger and longer lasting than normal. These results demonstrate that RK is required for normal rhodopsin deactivation.
    [Show full text]
  • Phosphorylation of G Protein-Coupled Receptors: from the Barcode Hypothesis to the Flute Model
    1521-0111/92/3/201–210$25.00 https://doi.org/10.1124/mol.116.107839 MOLECULAR PHARMACOLOGY Mol Pharmacol 92:201–210, September 2017 Copyright ª 2017 by The Author(s) This is an open access article distributed under the CC BY-NC Attribution 4.0 International license. MINIREVIEW—MOLECULAR PHARMACOLOGY IN CHINA Phosphorylation of G Protein-Coupled Receptors: From the Barcode Hypothesis to the Flute Model Zhao Yang, Fan Yang, Daolai Zhang, Zhixin Liu, Amy Lin, Chuan Liu, Peng Xiao, Xiao Yu, and Jin-Peng Sun Downloaded from Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People’s Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People’s Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.) Received December 10, 2016; accepted February 23, 2017 molpharm.aspetjournals.org ABSTRACT Seven transmembrane G protein-coupled receptors (GPCRs) distinct functional outcomes. Our recent work using unnatural are often phosphorylated at the C terminus and on intracellular amino acid incorporation and fluorine-19 nuclear magnetic loops in response to various extracellular stimuli. Phosphoryla- resonance (19F-NMR) spectroscopy led to the flute model, tion of GPCRs by GPCR kinases and certain other kinases which provides preliminary insight into the receptor phospho- can promote the recruitment of arrestin molecules. The arrestins coding mechanism, by which receptor phosphorylation pat- critically regulate GPCR functions not only by mediating terns are recognized by an array of phosphate-binding pockets receptor desensitization and internalization, but also by redi- on arrestin and are translated into distinct conformations.
    [Show full text]