101
|
Zu T, Guo S, Bardhi O, Ryskamp DA, Li J, Khoramian Tusi S, Engelbrecht A, Klippel K, Chakrabarty P, Nguyen L, Golde TE, Sonenberg N, Ranum LPW. Metformin inhibits RAN translation through PKR pathway and mitigates disease in C9orf72 ALS/FTD mice. Proc Natl Acad Sci U S A 2020; 117:18591-18599. [PMID: 32690681 PMCID: PMC7414156 DOI: 10.1073/pnas.2005748117] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Repeat associated non-AUG (RAN) translation is found in a growing number of microsatellite expansion diseases, but the mechanisms remain unclear. We show that RAN translation is highly regulated by the double-stranded RNA-dependent protein kinase (PKR). In cells, structured CAG, CCUG, CAGG, and G4C2 expansion RNAs activate PKR, which leads to increased levels of multiple RAN proteins. Blocking PKR using PKR-K296R, the TAR RNA binding protein or PKR-KO cells, reduces RAN protein levels. p-PKR is elevated in C9orf72 ALS/FTD human and mouse brains, and inhibiting PKR in C9orf72 BAC transgenic mice using AAV-PKR-K296R or the Food and Drug Administration (FDA)-approved drug metformin, decreases RAN proteins, and improves behavior and pathology. In summary, targeting PKR, including by use of metformin, is a promising therapeutic approach for C9orf72 ALS/FTD and other expansion diseases.
Collapse
Affiliation(s)
- Tao Zu
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
| | - Shu Guo
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
| | - Olgert Bardhi
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
| | - Daniel A Ryskamp
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
| | - Jian Li
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
| | - Solaleh Khoramian Tusi
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
| | - Avery Engelbrecht
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
| | - Kelena Klippel
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
| | - Paramita Chakrabarty
- Center for Translation Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Lien Nguyen
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
| | - Todd E Golde
- Center for Translation Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610
- McKnight Brain Institute, University of Florida, Gainesville, FL 32610
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montreal, A3A 1A3, Canada;
| | - Laura P W Ranum
- Center for NeuroGenetics, College of Medicine, University of Florida, Gainesville, FL 32610;
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610
- Genetics Institute, University of Florida, Gainesville, FL 32610
- McKnight Brain Institute, University of Florida, Gainesville, FL 32610
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610
- Norman Fixel Institute for Neurological Disease, University of Florida, Gainesville, FL 32608
| |
Collapse
|
102
|
He F, Flores BN, Krans A, Frazer M, Natla S, Niraula S, Adefioye O, Barmada SJ, Todd PK. The carboxyl termini of RAN translated GGGGCC nucleotide repeat expansions modulate toxicity in models of ALS/FTD. Acta Neuropathol Commun 2020; 8:122. [PMID: 32753055 PMCID: PMC7401224 DOI: 10.1186/s40478-020-01002-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
An intronic hexanucleotide repeat expansion in C9ORF72 causes familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This repeat is thought to elicit toxicity through RNA mediated protein sequestration and repeat-associated non-AUG (RAN) translation of dipeptide repeat proteins (DPRs). We generated a series of transgenic Drosophila models expressing GGGGCC (G4C2) repeats either inside of an artificial intron within a GFP reporter or within the 5' untranslated region (UTR) of GFP placed in different downstream reading frames. Expression of 484 intronic repeats elicited minimal alterations in eye morphology, viability, longevity, or larval crawling but did trigger RNA foci formation, consistent with prior reports. In contrast, insertion of repeats into the 5' UTR elicited differential toxicity that was dependent on the reading frame of GFP relative to the repeat. Greater toxicity correlated with a short and unstructured carboxyl terminus (C-terminus) in the glycine-arginine (GR) RAN protein reading frame. This change in C-terminal sequence triggered nuclear accumulation of all three RAN DPRs. A similar differential toxicity and dependence on the GR C-terminus was observed when repeats were expressed in rodent neurons. The presence of the native C-termini across all three reading frames was partly protective. Taken together, these findings suggest that C-terminal sequences outside of the repeat region may alter the behavior and toxicity of dipeptide repeat proteins derived from GGGGCC repeats.
Collapse
|
103
|
Bond S, Lopez-Lloreda C, Gannon PJ, Akay-Espinoza C, Jordan-Sciutto KL. The Integrated Stress Response and Phosphorylated Eukaryotic Initiation Factor 2α in Neurodegeneration. J Neuropathol Exp Neurol 2020; 79:123-143. [PMID: 31913484 DOI: 10.1093/jnen/nlz129] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
The proposed molecular mechanisms underlying neurodegenerative pathogenesis are varied, precluding the development of effective therapies for these increasingly prevalent disorders. One of the most consistent observations across neurodegenerative diseases is the phosphorylation of eukaryotic initiation factor 2α (eIF2α). eIF2α is a translation initiation factor, involved in cap-dependent protein translation, which when phosphorylated causes global translation attenuation. eIF2α phosphorylation is mediated by 4 kinases, which, together with their downstream signaling cascades, constitute the integrated stress response (ISR). While the ISR is activated by stresses commonly observed in neurodegeneration, such as oxidative stress, endoplasmic reticulum stress, and inflammation, it is a canonically adaptive signaling cascade. However, chronic activation of the ISR can contribute to neurodegenerative phenotypes such as neuronal death, memory impairments, and protein aggregation via apoptotic induction and other maladaptive outcomes downstream of phospho-eIF2α-mediated translation inhibition, including neuroinflammation and altered amyloidogenic processing, plausibly in a feed-forward manner. This review examines evidence that dysregulated eIF2a phosphorylation acts as a driver of neurodegeneration, including a survey of observations of ISR signaling in human disease, inspection of the overlap between ISR signaling and neurodegenerative phenomenon, and assessment of recent encouraging findings ameliorating neurodegeneration using developing pharmacological agents which target the ISR. In doing so, gaps in the field, including crosstalk of the ISR kinases and consideration of ISR signaling in nonneuronal central nervous system cell types, are highlighted.
Collapse
Affiliation(s)
- Sarah Bond
- From the Department of Biochemistry and Biophysics (SB); Department of Neuroscience (CL-L); Department of Pharmacology (PG), Perelman School of Medicine; Department of Basic and Translational Sciences (CA-E); and Department of Basic and Translational Sciences (KLJ-S), School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Claudia Lopez-Lloreda
- From the Department of Biochemistry and Biophysics (SB); Department of Neuroscience (CL-L); Department of Pharmacology (PG), Perelman School of Medicine; Department of Basic and Translational Sciences (CA-E); and Department of Basic and Translational Sciences (KLJ-S), School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patrick J Gannon
- From the Department of Biochemistry and Biophysics (SB); Department of Neuroscience (CL-L); Department of Pharmacology (PG), Perelman School of Medicine; Department of Basic and Translational Sciences (CA-E); and Department of Basic and Translational Sciences (KLJ-S), School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cagla Akay-Espinoza
- From the Department of Biochemistry and Biophysics (SB); Department of Neuroscience (CL-L); Department of Pharmacology (PG), Perelman School of Medicine; Department of Basic and Translational Sciences (CA-E); and Department of Basic and Translational Sciences (KLJ-S), School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kelly L Jordan-Sciutto
- From the Department of Biochemistry and Biophysics (SB); Department of Neuroscience (CL-L); Department of Pharmacology (PG), Perelman School of Medicine; Department of Basic and Translational Sciences (CA-E); and Department of Basic and Translational Sciences (KLJ-S), School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
104
|
Sun Y, Eshov A, Zhou J, Isiktas AU, Guo JU. C9orf72 arginine-rich dipeptide repeats inhibit UPF1-mediated RNA decay via translational repression. Nat Commun 2020; 11:3354. [PMID: 32620797 PMCID: PMC7335171 DOI: 10.1038/s41467-020-17129-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
Expansion of an intronic (GGGGCC)n repeat region within the C9orf72 gene is a main cause of familial amyotrophic lateral sclerosis and frontotemporal dementia (c9ALS/FTD). A hallmark of c9ALS/FTD is the accumulation of misprocessed RNAs, which are often targets of cellular RNA surveillance. Here, we show that RNA decay mechanisms involving upstream frameshift 1 (UPF1), including nonsense-mediated decay (NMD), are inhibited in c9ALS/FTD brains and in cultured cells expressing either of two arginine-rich dipeptide repeats (R-DPRs), poly(GR) and poly(PR). Mechanistically, although R-DPRs cause the recruitment of UPF1 to stress granules, stress granule formation is independent of NMD inhibition. Instead, NMD inhibition is primarily a result from global translational repression caused by R-DPRs. Overexpression of UPF1, but none of its NMD-deficient mutants, enhanced the survival of neurons treated by R-DPRs, suggesting that R-DPRs cause neurotoxicity in part by inhibiting cellular RNA surveillance. C9orf72 repeat expansion is the major genetic cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here, the authors show that transcriptome aberrations commonly found in c9ALS/FTD are a result from defects in cellular RNA surveillance pathways that involve an RNA helicase UPF1.
Collapse
Affiliation(s)
- Yu Sun
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Aziz Eshov
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jeffrey Zhou
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Atagun U Isiktas
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Junjie U Guo
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06520, USA. .,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
105
|
Tang X, Toro A, T G S, Gao J, Chalk J, Oskarsson B, Zhang K. Divergence, Convergence, and Therapeutic Implications: A Cell Biology Perspective of C9ORF72-ALS/FTD. Mol Neurodegener 2020; 15:34. [PMID: 32513219 PMCID: PMC7282082 DOI: 10.1186/s13024-020-00383-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Ever since a GGGGCC hexanucleotide repeat expansion mutation in C9ORF72 was identified as the most common cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), three competing but nonexclusive hypotheses to explain how this mutation causes diseases have been proposed and are still under debate. Recent studies in the field have tried to understand how the repeat expansion disrupts cellular physiology, which has suggested interesting convergence of these hypotheses on downstream, functional defects in cells, such as nucleocytoplasmic transport disruption, membrane-less organelle defects, and DNA damage. These studies have not only provided an integrated view of the disease mechanism but also revealed novel cell biology implicated in neurodegeneration. Furthermore, some of the discoveries have given rise to new ideas for therapeutic development. Here, we review the research progress on cellular pathophysiology of C9ORF72-mediated ALS and FTD and its therapeutic implication. We suggest that the repeat expansion drives pathogenesis through a combination of downstream defects, of which some can be therapeutic targets.
Collapse
Affiliation(s)
- Xiaoqiang Tang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Arturo Toro
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Sahana T G
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Junli Gao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Jessica Chalk
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Ke Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA. .,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA.
| |
Collapse
|
106
|
Kohata K, Miyoshi D. RNA phase separation-mediated direction of molecular trafficking under conditions of molecular crowding. Biophys Rev 2020; 12:669-676. [PMID: 32415614 DOI: 10.1007/s12551-020-00696-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Living cells are highly crowded with large and small biomolecules. The total concentration of biomolecules can reach 400 mg/ml, and 40% of the cell volume is occupied by biomolecules. Droplet formation in cells via liquid-liquid phase separation may play a role in controlling biochemical reactions in this complex molecular environment. Liquid-liquid phase separation generally involves nucleic acids and proteins as anionic and cationic components, respectively. Significant characteristics of droplets, which make them different from protein aggregation or fibril formation, are reversibility of formation and responsiveness to the molecular environment. In this review, we quantitatively describe the molecular environment inside cells and droplets that participate in controlling central dogma reactions. Finally, we discuss the importance of droplets under conditions of molecular crowding within living cells.
Collapse
Affiliation(s)
- Kazuki Kohata
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Daisuke Miyoshi
- Faculty of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
107
|
Costa-Mattioli M, Walter P. The integrated stress response: From mechanism to disease. Science 2020; 368:368/6489/eaat5314. [PMID: 32327570 DOI: 10.1126/science.aat5314] [Citation(s) in RCA: 721] [Impact Index Per Article: 180.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Protein quality control is essential for the proper function of cells and the organisms that they make up. The resulting loss of proteostasis, the processes by which the health of the cell's proteins is monitored and maintained at homeostasis, is associated with a wide range of age-related human diseases. Here, we highlight how the integrated stress response (ISR), a central signaling network that responds to proteostasis defects by tuning protein synthesis rates, impedes the formation of long-term memory. In addition, we address how dysregulated ISR signaling contributes to the pathogenesis of complex diseases, including cognitive disorders, neurodegeneration, cancer, diabetes, and metabolic disorders. The development of tools through which the ISR can be modulated promises to uncover new avenues to diminish pathologies resulting from it for clinical benefit.
Collapse
Affiliation(s)
- Mauro Costa-Mattioli
- Department of Neuroscience, Memory and Brain Research Center, Baylor College of Medicine, Houston, TX, USA.
| | - Peter Walter
- Howard Hughes Medical Institute and Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
108
|
McEachin ZT, Gendron TF, Raj N, García-Murias M, Banerjee A, Purcell RH, Ward PJ, Todd TW, Merritt-Garza ME, Jansen-West K, Hales CM, García-Sobrino T, Quintáns B, Holler CJ, Taylor G, San Millán B, Teijeira S, Yamashita T, Ohkubo R, Boulis NM, Xu C, Wen Z, Streichenberger N, Fogel BL, Kukar T, Abe K, Dickson DW, Arias M, Glass JD, Jiang J, Tansey MG, Sobrido MJ, Petrucelli L, Rossoll W, Bassell GJ. Chimeric Peptide Species Contribute to Divergent Dipeptide Repeat Pathology in c9ALS/FTD and SCA36. Neuron 2020; 107:292-305.e6. [PMID: 32375063 PMCID: PMC8138626 DOI: 10.1016/j.neuron.2020.04.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/11/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022]
Abstract
GGGGCC hexanucleotide repeat expansions (HREs) in C9orf72 cause amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) and lead to the production of aggregating dipeptide repeat proteins (DPRs) via repeat associated non-AUG (RAN) translation. Here, we show the similar intronic GGCCTG HREs that causes spinocerebellar ataxia type 36 (SCA36) is also translated into DPRs, including poly(GP) and poly(PR). We demonstrate that poly(GP) is more abundant in SCA36 compared to c9ALS/FTD patient tissue due to canonical AUG-mediated translation from intron-retained GGCCTG repeat RNAs. However, the frequency of the antisense RAN translation product poly(PR) is comparable between c9ALS/FTD and SCA36 patient samples. Interestingly, in SCA36 patient tissue, poly(GP) exists as a soluble species, and no TDP-43 pathology is present. We show that aggregate-prone chimeric DPR (cDPR) species underlie the divergent DPR pathology between c9ALS/FTD and SCA36. These findings reveal key differences in translation, solubility, and protein aggregation of DPRs between c9ALS/FTD and SCA36.
Collapse
Affiliation(s)
- Zachary T McEachin
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; Laboratory for Translational Cell Biology, Emory University, Atlanta, GA 30322, USA; Wallace H. Coulter Graduate Program in Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA.
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Nisha Raj
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; Laboratory for Translational Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - María García-Murias
- Centro de Investigación Biomédica en red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain; Neurogenetics Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario, SERGAS, Santiago de Compostela, Spain
| | - Anwesha Banerjee
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Ryan H Purcell
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; Laboratory for Translational Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Patricia J Ward
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Tiffany W Todd
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Karen Jansen-West
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chadwick M Hales
- Department of Neurology, Emory University, Atlanta, GA 30322, USA
| | - Tania García-Sobrino
- Department of Neurology, Hospital Clínico Universitario, SERGAS, Santiago de Compostela, Spain
| | - Beatriz Quintáns
- Centro de Investigación Biomédica en red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain; Neurogenetics Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario, SERGAS, Santiago de Compostela, Spain
| | - Christopher J Holler
- Department of Pharmacology & Chemical Biology, Emory University, Atlanta, GA 30322, USA
| | - Georgia Taylor
- Department of Pharmacology & Chemical Biology, Emory University, Atlanta, GA 30322, USA
| | - Beatriz San Millán
- Rare Diseases and Pediatric Medicine Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Pathology Department, Complexo Hospitalario Universitario de Vigo (CHUVI), SERGAS, Vigo, Spain
| | - Susana Teijeira
- Rare Diseases and Pediatric Medicine Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Pathology Department, Complexo Hospitalario Universitario de Vigo (CHUVI), SERGAS, Vigo, Spain
| | - Toru Yamashita
- Department of Neurology, Okayama University, Okayama, Japan
| | - Ryuichi Ohkubo
- Department of Neurology, Fujimoto General Hospital, Miyazaki, Japan
| | - Nicholas M Boulis
- Department of Neurosurgery, Emory University, Atlanta, GA 30322, USA
| | - Chongchong Xu
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA 30322, USA
| | - Zhexing Wen
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; Laboratory for Translational Cell Biology, Emory University, Atlanta, GA 30322, USA; Department of Neurology, Emory University, Atlanta, GA 30322, USA; Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA 30322, USA
| | - Nathalie Streichenberger
- Hospices Civils de Lyon, Lyon, France; Université Claude Bernard Lyon, Lyon, France; Institut NeuroMyogène CNRS UMR 5310
| | | | - Brent L Fogel
- Department of Neurology & Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Thomas Kukar
- Department of Neurology, Emory University, Atlanta, GA 30322, USA; Department of Pharmacology & Chemical Biology, Emory University, Atlanta, GA 30322, USA
| | - Koji Abe
- Department of Neurology, Okayama University, Okayama, Japan
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Manuel Arias
- Neurogenetics Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario, SERGAS, Santiago de Compostela, Spain; Department of Neurology, Hospital Clínico Universitario, SERGAS, Santiago de Compostela, Spain
| | - Jonathan D Glass
- Department of Neurology, Emory University, Atlanta, GA 30322, USA
| | - Jie Jiang
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Malú G Tansey
- Department of Neuroscience, University of Florida, Gainesville, FL 32607, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32607, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32607, USA
| | - María-Jesús Sobrido
- Centro de Investigación Biomédica en red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain; Neurogenetics Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario, SERGAS, Santiago de Compostela, Spain
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Wilfried Rossoll
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; Laboratory for Translational Cell Biology, Emory University, Atlanta, GA 30322, USA; Wallace H. Coulter Graduate Program in Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA 30332, USA; Department of Neurology, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
109
|
Systematic microsatellite repeat expansion cloning and validation. Hum Genet 2020; 139:1233-1246. [PMID: 32277284 DOI: 10.1007/s00439-020-02165-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/04/2020] [Indexed: 10/24/2022]
Abstract
Approximately 3% of the human genome is composed of short tandem repeat (STR) DNA sequence known as microsatellites, which can be found in both coding and non-coding regions. When associated with genic regions, expansion of microsatellite repeats beyond a critical threshold causes dozens of neurological repeat expansion disorders. To better understand the molecular pathology of repeat expansion disorders, precise cloning of microsatellite repeat sequence and expansion size is highly valuable. Unfortunately, cloning repeat expansions is often challenging and presents a significant bottleneck to practical investigation. Here, we describe a clear method for seamless and systematic cloning of practically any microsatellite repeat expansion. We use cloning and expansion of GGGGCC repeats, which are the leading genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), as an example. We employ a recursive directional ligation (RDL) technique to build multiple GGGGCC repeat-containing vectors. We describe methods to validate repeat expansion cloning, including diagnostic restriction digestion, PCR across the repeat, and next-generation long-read MinION nanopore sequencing. Validated cloning of microsatellite repeats beyond the critical expansion threshold can facilitate step-by-step characterization of disease mechanisms at the cellular and molecular level.
Collapse
|
110
|
Rudich P, Watkins S, Lamitina T. PolyQ-independent toxicity associated with novel translational products from CAG repeat expansions. PLoS One 2020; 15:e0227464. [PMID: 32240172 PMCID: PMC7117740 DOI: 10.1371/journal.pone.0227464] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/11/2020] [Indexed: 02/08/2023] Open
Abstract
Expanded CAG nucleotide repeats are the underlying genetic cause of at least 14 incurable diseases, including Huntington’s disease (HD). The toxicity associated with many CAG repeat expansions is thought to be due to the translation of the CAG repeat to create a polyQ protein, which forms toxic oligomers and aggregates. However, recent studies show that HD CAG repeats undergo a non-canonical form of translation called Repeat-associated non-AUG dependent (RAN) translation. RAN translation of the CAG sense and CUG anti-sense RNAs produces six distinct repeat peptides: polyalanine (polyAla, from both CAG and CUG repeats), polyserine (polySer), polyleucine (polyLeu), polycysteine (polyCys), and polyglutamine (polyGln). The toxic potential of individual CAG-derived RAN polypeptides is not well understood. We developed pure C. elegans protein models for each CAG RAN polypeptide using codon-varied expression constructs that preserve RAN protein sequence but eliminate repetitive CAG/CUG RNA. While all RAN polypeptides formed aggregates, only polyLeu was consistently toxic across multiple cell types. In GABAergic neurons, which exhibit significant neurodegeneration in HD patients, codon-varied (Leu)38, but not (Gln)38, caused substantial neurodegeneration and motility defects. Our studies provide the first in vivo evaluation of CAG-derived RAN polypeptides in a multicellular model organism and suggest that polyQ-independent mechanisms, such as RAN-translated polyLeu peptides, may have a significant pathological role in CAG repeat expansion disorders.
Collapse
Affiliation(s)
- Paige Rudich
- Graduate Program in Cell Biology and Molecular Physiology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States of America
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Simon Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Todd Lamitina
- Graduate Program in Cell Biology and Molecular Physiology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States of America
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
- Division of Child Neurology, Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
111
|
Abstract
The (GGGGCC)n repeat expansion in C9orf72, which is the most common cause of frontotemporal dementia and amyotrophic lateral sclerosis, is translated through repeat-associated non-AUG (RAN) translation. In this issue of Neuron, Cheng et al. (2019) report that the helicase DDX3X, which unwinds (or relaxes) RNA, suppresses RAN translation and toxicity.
Collapse
Affiliation(s)
- Katherine M Wilson
- UK Dementia Research Institute at UCL, Gower Street, London WC1E 6BT, UK; Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London WC1N 3BG, UK
| | - Bhavana Muralidharan
- UK Dementia Research Institute at UCL, Gower Street, London WC1E 6BT, UK; Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London WC1N 3BG, UK; Brain Development and Disease Mechanisms, Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India, 560065
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL, Gower Street, London WC1E 6BT, UK; Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London WC1N 3BG, UK.
| |
Collapse
|
112
|
Rodriguez CM, Wright SE, Kearse MG, Haenfler JM, Flores BN, Liu Y, Ifrim MF, Glineburg MR, Krans A, Jafar-Nejad P, Sutton MA, Bassell GJ, Parent JM, Rigo F, Barmada SJ, Todd PK. A native function for RAN translation and CGG repeats in regulating fragile X protein synthesis. Nat Neurosci 2020; 23:386-397. [PMID: 32066985 PMCID: PMC7668390 DOI: 10.1038/s41593-020-0590-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
Abstract
Repeat-associated non-AUG-initiated translation of expanded CGG repeats (CGG RAN) from the FMR1 5'-leader produces toxic proteins that contribute to neurodegeneration in fragile X-associated tremor/ataxia syndrome. Here we describe how unexpanded CGG repeats and their translation play conserved roles in regulating fragile X protein (FMRP) synthesis. In neurons, CGG RAN acts as an inhibitory upstream open reading frame to suppress basal FMRP production. Activation of mGluR5 receptors enhances FMRP synthesis. This enhancement requires both the CGG repeat and CGG RAN initiation sites. Using non-cleaving antisense oligonucleotides (ASOs), we selectively blocked CGG RAN. This ASO blockade enhanced endogenous FMRP expression in human neurons. In human and rodent neurons, CGG RAN-blocking ASOs suppressed repeat toxicity and prolonged survival. These findings delineate a native function for CGG repeats and RAN translation in regulating basal and activity-dependent FMRP synthesis, and they demonstrate the therapeutic potential of modulating CGG RAN translation in fragile X-associated disorders.
Collapse
Affiliation(s)
- Caitlin M Rodriguez
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Shannon E Wright
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Michael G Kearse
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Department of Biological Chemistry and Pharmacology, Center for RNA Biology, Ohio State University, Columbus, OH, USA
| | - Jill M Haenfler
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Brittany N Flores
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Yu Liu
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Marius F Ifrim
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mary R Glineburg
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Amy Krans
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | | | - Michael A Sutton
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jack M Parent
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | - Sami J Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Peter K Todd
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| |
Collapse
|
113
|
Friedman-Gohas M, Elizur SE, Dratviman-Storobinsky O, Aizer A, Haas J, Raanani H, Orvieto R, Cohen Y. FMRpolyG accumulates in FMR1 premutation granulosa cells. J Ovarian Res 2020; 13:22. [PMID: 32101156 PMCID: PMC7045455 DOI: 10.1186/s13048-020-00623-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/10/2020] [Indexed: 12/18/2022] Open
Abstract
Background Fragile X premutation (Amplification of CGG number 55–200) is associated with increased risk for fragile X-Associated Premature Ovarian Insufficiency (FXPOI) in females and fragile X-associated tremor/ataxia syndrome (FXTAS) predominantly in males. Recently, it has been shown that CGG repeats trigger repeat associated non-AUG initiated translation (RAN) of a cryptic polyglycine-containing protein, FMRpolyG. This protein accumulates in ubiquitin-positive inclusions in neuronal brain cells of FXTAS patients and may lead to protein-mediated neurodegeneration. FMRpolyG inclusions were also found in ovary stromal cells of a FXPOI patient. The role of FMRpolyG expression has not been thoroughly examined in folliculogenesis related cells. The main goal of this study is to evaluate whether FMRpolyG accumulates in mural granulosa cells of FMR1 premutation carriers. Following FMRpolyG detection, we aim to examine premutation transfected COV434 as a suitable model used to identify RAN translation functions in FXPOI pathogenesis. Results FMRpolyG and ubiquitin immunostained mural granulosa cells from six FMR1 premutation carriers demonstrated FMRpolyG aggregates. However, co-localization of FMRpolyG and ubiquitin appeared to vary within the FMR1 premutation carriers’ group as three exhibited partial ubiquitin and FMRpolyG double staining and three premutation carriers demonstrated FMRpolyG single staining. None of the granulosa cells from the five control women expressed FMRpolyG. Additionally, human ovarian granulosa tumor, COV434, were transfected with two plasmids; both expressing 99CGG repeats but only one enables FMRpolyG expression. Like in granulosa cells from FMR1 premutation carriers, FMRpolyG aggregates were found only in COV434 transfected with expended CGG repeats and the ability to express FMRpolyG. Conclusions Corresponding with previous studies in FXTAS, we demonstrated accumulation of FMRpolyG in mural granulosa cells of FMR1 premutation carriers. We also suggest that following further investigation, the premutation transfected COV434 might be an appropriate model for RAN translation studies. Detecting FMRpolyG accumulation in folliculogenesis related cells supports previous observations and imply a possible common protein-mediated toxic mechanism for both FXPOI and FXTAS.
Collapse
Affiliation(s)
- M Friedman-Gohas
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - S E Elizur
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,IVF Unit, Chaim Sheba Medical Centre, Tel-Hashomer, 52621, Ramat-Gan, Israel
| | - O Dratviman-Storobinsky
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,IVF Unit, Chaim Sheba Medical Centre, Tel-Hashomer, 52621, Ramat-Gan, Israel
| | - A Aizer
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,IVF Unit, Chaim Sheba Medical Centre, Tel-Hashomer, 52621, Ramat-Gan, Israel
| | - J Haas
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,IVF Unit, Chaim Sheba Medical Centre, Tel-Hashomer, 52621, Ramat-Gan, Israel
| | - H Raanani
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,IVF Unit, Chaim Sheba Medical Centre, Tel-Hashomer, 52621, Ramat-Gan, Israel
| | - R Orvieto
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,IVF Unit, Chaim Sheba Medical Centre, Tel-Hashomer, 52621, Ramat-Gan, Israel
| | - Y Cohen
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel. .,IVF Unit, Chaim Sheba Medical Centre, Tel-Hashomer, 52621, Ramat-Gan, Israel.
| |
Collapse
|
114
|
Boivin M, Pfister V, Gaucherot A, Ruffenach F, Negroni L, Sellier C, Charlet-Berguerand N. Reduced autophagy upon C9ORF72 loss synergizes with dipeptide repeat protein toxicity in G4C2 repeat expansion disorders. EMBO J 2020; 39:e100574. [PMID: 31930538 PMCID: PMC7024836 DOI: 10.15252/embj.2018100574] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/28/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022] Open
Abstract
Expansion of G4C2 repeats within the C9ORF72 gene is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Such repeats lead to decreased expression of the autophagy regulator C9ORF72 protein. Furthermore, sense and antisense repeats are translated into toxic dipeptide repeat (DPR) proteins. It is unclear how these repeats are translated, and in which way their translation and the reduced expression of C9ORF72 modulate repeat toxicity. Here, we found that sense and antisense repeats are translated upon initiation at canonical AUG or near‐cognate start codons, resulting in polyGA‐, polyPG‐, and to a lesser degree polyGR‐DPR proteins. However, accumulation of these proteins is prevented by autophagy. Importantly, reduced C9ORF72 levels lead to suboptimal autophagy, thereby impairing clearance of DPR proteins and causing their toxic accumulation, ultimately resulting in neuronal cell death. Of clinical importance, pharmacological compounds activating autophagy can prevent neuronal cell death caused by DPR proteins accumulation. These results suggest the existence of a double‐hit pathogenic mechanism in ALS/FTD, whereby reduced expression of C9ORF72 synergizes with DPR protein accumulation and toxicity.
Collapse
Affiliation(s)
- Manon Boivin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
| | - Véronique Pfister
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
| | - Angeline Gaucherot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
| | - Frank Ruffenach
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
| | - Luc Negroni
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
| | - Chantal Sellier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
| | - Nicolas Charlet-Berguerand
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
| |
Collapse
|
115
|
Nguyen L, Montrasio F, Pattamatta A, Tusi SK, Bardhi O, Meyer KD, Hayes L, Nakamura K, Banez-Coronel M, Coyne A, Guo S, Laboissonniere LA, Gu Y, Narayanan S, Smith B, Nitsch RM, Kankel MW, Rushe M, Rothstein J, Zu T, Grimm J, Ranum LPW. Antibody Therapy Targeting RAN Proteins Rescues C9 ALS/FTD Phenotypes in C9orf72 Mouse Model. Neuron 2019; 105:645-662.e11. [PMID: 31831332 DOI: 10.1016/j.neuron.2019.11.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/10/2019] [Accepted: 11/05/2019] [Indexed: 10/25/2022]
Abstract
The intronic C9orf72 G4C2 expansion, the most common genetic cause of ALS and FTD, produces sense- and antisense-expansion RNAs and six dipeptide repeat-associated, non-ATG (RAN) proteins, but their roles in disease are unclear. We generated high-affinity human antibodies targeting GA or GP RAN proteins. These antibodies cross the blood-brain barrier and co-localize with intracellular RAN aggregates in C9-ALS/FTD BAC mice. In cells, α-GA1 interacts with TRIM21, and α-GA1 treatment reduced GA levels, increased GA turnover, and decreased RAN toxicity and co-aggregation of proteasome and autophagy proteins to GA aggregates. In C9-BAC mice, α-GA1 reduced GA as well as GP and GR proteins, improved behavioral deficits, decreased neuroinflammation and neurodegeneration, and increased survival. Glycosylation of the Fc region of α-GA1 is important for cell entry and efficacy. These data demonstrate that RAN proteins drive C9-ALS/FTD in C9-BAC transgenic mice and establish a novel therapeutic approach for C9orf72 ALS/FTD and other RAN-protein diseases.
Collapse
Affiliation(s)
- Lien Nguyen
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | | | - Amrutha Pattamatta
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Solaleh Khoramian Tusi
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Olgert Bardhi
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Kevin D Meyer
- Neurimmune AG, 8952 Schlieren, Switzerland; Institute for Regenerative Medicine-IREM, University of Zurich, 8952 Schlieren, Switzerland
| | - Lindsey Hayes
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Katsuya Nakamura
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Monica Banez-Coronel
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Alyssa Coyne
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Shu Guo
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Lauren A Laboissonniere
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Yuanzheng Gu
- Neuromuscular and Movement Disorders, Biogen, Cambridge, MA 02142, USA
| | | | - Benjamin Smith
- Neuromuscular and Movement Disorders, Biogen, Cambridge, MA 02142, USA
| | - Roger M Nitsch
- Neurimmune AG, 8952 Schlieren, Switzerland; Institute for Regenerative Medicine-IREM, University of Zurich, 8952 Schlieren, Switzerland
| | - Mark W Kankel
- Neuromuscular and Movement Disorders, Biogen, Cambridge, MA 02142, USA
| | - Mia Rushe
- Neuromuscular and Movement Disorders, Biogen, Cambridge, MA 02142, USA
| | - Jeffrey Rothstein
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Tao Zu
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA
| | - Jan Grimm
- Neurimmune AG, 8952 Schlieren, Switzerland
| | - Laura P W Ranum
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
116
|
Production of poly(GA) in C9ORF72 patient motor neurons derived from induced pluripotent stem cells. Acta Neuropathol 2019; 138:1099-1101. [PMID: 31624870 PMCID: PMC6851345 DOI: 10.1007/s00401-019-02083-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 11/17/2022]
|
117
|
Cook C, Petrucelli L. Genetic Convergence Brings Clarity to the Enigmatic Red Line in ALS. Neuron 2019; 101:1057-1069. [PMID: 30897357 DOI: 10.1016/j.neuron.2019.02.032] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/07/2019] [Accepted: 02/19/2019] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an aggressive neurodegenerative disorder that orchestrates an attack on the motor nervous system that is unrelenting. Recent discoveries into the pathogenic consequences of repeat expansions in C9ORF72, which are the most common genetic cause of ALS, combined with the identification of new genetic mutations are providing novel insight into the underlying mechanism(s) that cause ALS. In particular, the myriad of functions linked to ALS-associated genes have collectively implicated four main pathways in disease pathogenesis, including RNA metabolism and translational biology; protein quality control; cytoskeletal integrity and trafficking; and mitochondrial function and transport. Through the identification of common disease mechanisms on which multiple ALS genes converge, key targets for potential therapeutic intervention are highlighted.
Collapse
Affiliation(s)
- Casey Cook
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neurobiology of Disease Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Neurobiology of Disease Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA.
| |
Collapse
|
118
|
Abstract
Recent advances suggest that the response of RNA metabolism to stress has an important role in the pathophysiology of neurodegenerative diseases, particularly amyotrophic lateral sclerosis, frontotemporal dementias and Alzheimer disease. RNA-binding proteins (RBPs) control the utilization of mRNA during stress, in part through the formation of membraneless organelles termed stress granules (SGs). These structures form through a process of liquid-liquid phase separation. Multiple biochemical pathways regulate SG biology. The major signalling pathways regulating SG formation include the mammalian target of rapamycin (mTOR)-eukaryotic translation initiation factor 4F (eIF4F) and eIF2α pathways, whereas the pathways regulating SG dispersion and removal are mediated by valosin-containing protein and the autolysosomal cascade. Post-translational modifications of RBPs also strongly contribute to the regulation of SGs. Evidence indicates that SGs are supposed to be transient structures, but the chronic stresses associated with ageing lead to chronic, persistent SGs that appear to act as a nidus for the aggregation of disease-related proteins. We suggest a model describing how intrinsic vulnerabilities within the cellular RNA metabolism might lead to the pathological aggregation of RBPs when SGs become persistent. This process might accelerate the pathophysiology of many neurodegenerative diseases and myopathies, and it suggests new targets for disease intervention.
Collapse
Affiliation(s)
- Benjamin Wolozin
- Department of Pharmacology, Boston University School of Medicine, Boston, MA, USA.
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.
| | - Pavel Ivanov
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| |
Collapse
|
119
|
Hui KK, Chen YK, Endo R, Tanaka M. Translation from the Ribosome to the Clinic: Implication in Neurological Disorders and New Perspectives from Recent Advances. Biomolecules 2019; 9:E680. [PMID: 31683805 PMCID: PMC6920867 DOI: 10.3390/biom9110680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/27/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
De novo protein synthesis by the ribosome and its multitude of co-factors must occur in a tightly regulated manner to ensure that the correct proteins are produced accurately at the right time and, in some cases, also in the proper location. With novel techniques such as ribosome profiling and cryogenic electron microscopy, our understanding of this basic biological process is better than ever and continues to grow. Concurrently, increasing attention is focused on how translational regulation in the brain may be disrupted during the progression of various neurological disorders. In fact, translational dysregulation is now recognized as the de facto pathogenic cause for some disorders. Novel mechanisms including ribosome stalling, ribosome-associated quality control, and liquid-liquid phase separation are closely linked to translational regulation, and may thus be involved in the pathogenic process. The relationships between translational dysregulation and neurological disorders, as well as the ways through which we may be able to reverse those detrimental effects, will be examined in this review.
Collapse
Affiliation(s)
- Kelvin K Hui
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Yi-Kai Chen
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Ryo Endo
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
120
|
Green KM, Sheth UJ, Flores BN, Wright SE, Sutter AB, Kearse MG, Barmada SJ, Ivanova MI, Todd PK. High-throughput screening yields several small-molecule inhibitors of repeat-associated non-AUG translation. J Biol Chem 2019; 294:18624-18638. [PMID: 31649034 DOI: 10.1074/jbc.ra119.009951] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/16/2019] [Indexed: 12/18/2022] Open
Abstract
Repeat-associated non-AUG (RAN) translation is a noncanonical translation initiation event that occurs at nucleotide-repeat expansion mutations that are associated with several neurodegenerative diseases, including fragile X-associated tremor ataxia syndrome (FXTAS), ALS, and frontotemporal dementia (FTD). Translation of expanded repeats produces toxic proteins that accumulate in human brains and contribute to disease pathogenesis. Consequently, RAN translation constitutes a potentially important therapeutic target for managing multiple neurodegenerative disorders. Here, we adapted a previously developed RAN translation assay to a high-throughput format to screen 3,253 bioactive compounds for inhibition of RAN translation of expanded CGG repeats associated with FXTAS. We identified five diverse small molecules that dose-dependently inhibited CGG RAN translation, while relatively sparing canonical translation. All five compounds also inhibited RAN translation of expanded GGGGCC repeats associated with ALS and FTD. Using CD and native gel analyses, we found evidence that three of these compounds, BIX01294, CP-31398, and propidium iodide, bind directly to the repeat RNAs. These findings provide proof-of-principle supporting the development of selective small-molecule RAN translation inhibitors that act across multiple disease-causing repeats.
Collapse
Affiliation(s)
- Katelyn M Green
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109; Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan 48109
| | - Udit J Sheth
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Brittany N Flores
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109; Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan 48109
| | - Shannon E Wright
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Alexandra B Sutter
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109
| | - Michael G Kearse
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109; Department of Biological Chemistry and Pharmacology, Center for RNA Biology, Ohio State University, Columbus, Ohio 43210
| | - Sami J Barmada
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109; Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan 48109
| | - Magdalena I Ivanova
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109; Biophysics Program, University of Michigan, Ann Arbor, Michigan 48109
| | - Peter K Todd
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109; Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan 48109; Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan 48105.
| |
Collapse
|
121
|
Baradaran-Heravi Y, Van Broeckhoven C, van der Zee J. Stress granule mediated protein aggregation and underlying gene defects in the FTD-ALS spectrum. Neurobiol Dis 2019; 134:104639. [PMID: 31626953 DOI: 10.1016/j.nbd.2019.104639] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 09/12/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Stress granules (SGs) are dynamic membraneless compartments composed out of RNA-binding proteins (RBPs) and RNA molecules that assemble temporarily to allow the cell to cope with cellular stress by stalling mRNA translation and moving synthesis towards cytoprotective proteins. Aberrant SGs have become prime suspects in the nucleation of toxic protein aggregation in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Perturbed SG dynamics appears to be mediated by alterations in RNA binding proteins (RBP). Indeed, a growing number of FTD and/or ALS related RBPs coding genes (TDP43, FUS, EWSR1, TAF15, hnRNPA1, hnRNPA2B1, ATXN2, TIA1) have been identified to interfere with SG formation through mutation of their low-complexity domain (LCD), and thereby cause or influence disease. Interestingly, disease pathways associated to the C9orf72 repeat expansion, the leading genetic cause of the FTD-ALS spectrum, intersect with SG-mediated protein aggregate formation. In this review, we provide a comprehensive overview of known SG proteins and their genetic contribution to the FTD-ALS spectrum. Importantly, multiple LCD-baring RBPs have already been identified in FTD-ALS that have not yet been genetically linked to disease. These should be considered candidate genes and offer opportunities for gene prioritization when mining sequencing data of unresolved FTD and ALS. Further, we zoom into the current understanding of the molecular processes of perturbed RBP function leading to disturbed SG dynamics, RNA metabolism, and pathological inclusions. Finally, we indicate how these gained insights open new avenues for therapeutic strategies targeting phase separation and SG dynamics to reverse pathological protein aggregation and protect against toxicity.
Collapse
Affiliation(s)
- Yalda Baradaran-Heravi
- Neurodegenerative Brain Diseases group, Center for Molecular Neurology, VIB, Antwerp, Belgium; Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases group, Center for Molecular Neurology, VIB, Antwerp, Belgium; Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.
| | - Julie van der Zee
- Neurodegenerative Brain Diseases group, Center for Molecular Neurology, VIB, Antwerp, Belgium; Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
122
|
Cheng W, Wang S, Zhang Z, Morgens DW, Hayes LR, Lee S, Portz B, Xie Y, Nguyen BV, Haney MS, Yan S, Dong D, Coyne AN, Yang J, Xian F, Cleveland DW, Qiu Z, Rothstein JD, Shorter J, Gao FB, Bassik MC, Sun S. CRISPR-Cas9 Screens Identify the RNA Helicase DDX3X as a Repressor of C9ORF72 (GGGGCC)n Repeat-Associated Non-AUG Translation. Neuron 2019; 104:885-898.e8. [PMID: 31587919 DOI: 10.1016/j.neuron.2019.09.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/16/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022]
Abstract
Hexanucleotide GGGGCC repeat expansion in C9ORF72 is the most prevalent genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). One pathogenic mechanism is the aberrant accumulation of dipeptide repeat (DPR) proteins produced by the unconventional translation of expanded RNA repeats. Here, we performed genome-wide CRISPR-Cas9 screens for modifiers of DPR protein production in human cells. We found that DDX3X, an RNA helicase, suppresses the repeat-associated non-AUG translation of GGGGCC repeats. DDX3X directly binds to (GGGGCC)n RNAs but not antisense (CCCCGG)n RNAs. Its helicase activity is essential for the translation repression. Reduction of DDX3X increases DPR levels in C9ORF72-ALS/FTD patient cells and enhances (GGGGCC)n-mediated toxicity in Drosophila. Elevating DDX3X expression is sufficient to decrease DPR levels, rescue nucleocytoplasmic transport abnormalities, and improve survival of patient iPSC-differentiated neurons. This work identifies genetic modifiers of DPR protein production and provides potential therapeutic targets for C9ORF72-ALS/FTD.
Collapse
Affiliation(s)
- Weiwei Cheng
- Department of Pathology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shaopeng Wang
- Department of Pathology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhe Zhang
- Department of Pathology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David W Morgens
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lindsey R Hayes
- Brain Science Institute and Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Soojin Lee
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Bede Portz
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yongzhi Xie
- Department of Pathology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Baotram V Nguyen
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael S Haney
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shirui Yan
- Department of Pathology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daoyuan Dong
- Department of Pathology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alyssa N Coyne
- Brain Science Institute and Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Junhua Yang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Fengfan Xian
- Department of Pathology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Don W Cleveland
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Zhaozhu Qiu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeffrey D Rothstein
- Brain Science Institute and Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shuying Sun
- Department of Pathology and Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
123
|
Jiang J, Ravits J. Pathogenic Mechanisms and Therapy Development for C9orf72 Amyotrophic Lateral Sclerosis/Frontotemporal Dementia. Neurotherapeutics 2019; 16:1115-1132. [PMID: 31667754 PMCID: PMC6985338 DOI: 10.1007/s13311-019-00797-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In 2011, a hexanucleotide repeat expansion in the first intron of the C9orf72 gene was identified as the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The proposed disease mechanisms include loss of C9orf72 function and gain of toxicity from the bidirectionally transcribed repeat-containing RNAs. Over the last few years, substantial progress has been made to determine the contribution of loss and gain of function in disease pathogenesis. The extensive body of molecular, cellular, animal, and human neuropathological studies is conflicted, but the predominance of evidence favors gain of toxicity as the main pathogenic mechanism for C9orf72 repeat expansions. Alterations in several downstream cellular functions, such as nucleocytoplasmic transport and autophagy, are implicated. Exciting progress has also been made in therapy development targeting this mutation, such as by antisense oligonucleotide therapies targeting sense transcripts and small molecules targeting nucleocytoplasmic transport, and these are now in phase 1 clinical trials.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Cell Biology, Emory University, Atlanta, GA, 30322, USA.
| | - John Ravits
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
124
|
Goodman LD, Bonini NM. Repeat-associated non-AUG (RAN) translation mechanisms are running into focus for GGGGCC-repeat associated ALS/FTD. Prog Neurobiol 2019; 183:101697. [PMID: 31550516 DOI: 10.1016/j.pneurobio.2019.101697] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/31/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022]
Abstract
Many human diseases are associated with the expansion of repeat sequences within the genes. It has become clear that expressed disease transcripts bearing such long repeats can undergo translation, even in the absence of a canonical AUG start codon. Termed "RAN translation" for repeat associated non-AUG translation, this process is becoming increasingly prominent as a contributor to these disorders. Here we discuss mechanisms and variables that impact translation of the repeat sequences associated with the C9orf72 gene. Expansions of a G4C2 repeat within intron 1 of this gene are associated with the motor neuron disease ALS and dementia FTD, which comprise a clinical and pathological spectrum. RAN translation of G4C2 repeat expansions has been studied in cells in culture (ex vivo) and in the fly in vivo. Cellular states that lead to RAN translation, like stress, may be critical contributors to disease progression. Greater elucidation of the mechanisms that impact this process and the factors contributing will lead to greater understanding of the repeat expansion diseases, to the potential development of novel approaches to therapeutics, and to a greater understanding of how these players impact biological processes in the absence of disease.
Collapse
Affiliation(s)
- Lindsey D Goodman
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy M Bonini
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
125
|
Yamada SB, Gendron TF, Niccoli T, Genuth NR, Grosely R, Shi Y, Glaria I, Kramer NJ, Nakayama L, Fang S, Dinger TJI, Thoeng A, Rocha G, Barna M, Puglisi JD, Partridge L, Ichida JK, Isaacs AM, Petrucelli L, Gitler AD. RPS25 is required for efficient RAN translation of C9orf72 and other neurodegenerative disease-associated nucleotide repeats. Nat Neurosci 2019; 22:1383-1388. [PMID: 31358992 PMCID: PMC6713615 DOI: 10.1038/s41593-019-0455-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 06/20/2019] [Indexed: 12/18/2022]
Abstract
Nucleotide repeat expansions in the C9orf72 gene are the most common cause of amyotrophic lateral sclerosis and frontotemporal dementia. Unconventional translation (RAN translation) of C9orf72 repeats generates dipeptide repeat proteins that can cause neurodegeneration. We performed a genetic screen for regulators of RAN translation and identified small ribosomal protein subunit 25 (RPS25), presenting a potential therapeutic target for C9orf72-related amyotrophic lateral sclerosis and frontotemporal dementia and other neurodegenerative diseases caused by nucleotide repeat expansions.
Collapse
Affiliation(s)
- Shizuka B Yamada
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Teresa Niccoli
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, London, UK
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| | - Naomi R Genuth
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Rosslyn Grosely
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yingxiao Shi
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Idoia Glaria
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, London, UK
| | - Nicholas J Kramer
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Lisa Nakayama
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Shirleen Fang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Tai J I Dinger
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Annora Thoeng
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, London, UK
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| | - Gabriel Rocha
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Maria Barna
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph D Puglisi
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Linda Partridge
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, London, UK
| | | | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
126
|
Linsalata AE, He F, Malik AM, Glineburg MR, Green KM, Natla S, Flores BN, Krans A, Archbold HC, Fedak SJ, Barmada SJ, Todd PK. DDX3X and specific initiation factors modulate FMR1 repeat-associated non-AUG-initiated translation. EMBO Rep 2019; 20:e47498. [PMID: 31347257 PMCID: PMC6726903 DOI: 10.15252/embr.201847498] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/19/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
A CGG trinucleotide repeat expansion in the 5' UTR of FMR1 causes the neurodegenerative disorder Fragile X-associated tremor/ataxia syndrome (FXTAS). This repeat supports a non-canonical mode of protein synthesis known as repeat-associated, non-AUG (RAN) translation. The mechanism underlying RAN translation at CGG repeats remains unclear. To identify modifiers of RAN translation and potential therapeutic targets, we performed a candidate-based screen of eukaryotic initiation factors and RNA helicases in cell-based assays and a Drosophila melanogaster model of FXTAS. We identified multiple modifiers of toxicity and RAN translation from an expanded CGG repeat in the context of the FMR1 5'UTR. These include the DEAD-box RNA helicase belle/DDX3X, the helicase accessory factors EIF4B/4H, and the start codon selectivity factors EIF1 and EIF5. Disrupting belle/DDX3X selectively inhibited FMR1 RAN translation in Drosophila in vivo and cultured human cells, and mitigated repeat-induced toxicity in Drosophila and primary rodent neurons. These findings implicate RNA secondary structure and start codon fidelity as critical elements mediating FMR1 RAN translation and identify potential targets for treating repeat-associated neurodegeneration.
Collapse
Affiliation(s)
- Alexander E Linsalata
- Cellular and Molecular Biology Graduate ProgramUniversity of MichiganAnn ArborMIUSA
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Fang He
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
- Department of Biological and Health SciencesTexas A&M University, KingsvilleKingsvilleTXUSA
| | - Ahmed M Malik
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
- Neuroscience Graduate ProgramUniversity of MichiganAnn ArborMIUSA
| | | | - Katelyn M Green
- Cellular and Molecular Biology Graduate ProgramUniversity of MichiganAnn ArborMIUSA
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Sam Natla
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Brittany N Flores
- Cellular and Molecular Biology Graduate ProgramUniversity of MichiganAnn ArborMIUSA
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Amy Krans
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | | | | | - Sami J Barmada
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
| | - Peter K Todd
- Department of NeurologyUniversity of MichiganAnn ArborMIUSA
- Ann Arbor VA Medical CenterAnn ArborMIUSA
| |
Collapse
|
127
|
Abstract
The discovery that repeat expansions in the C9orf72 gene are a frequent cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) has revolutionized our understanding of these diseases. Substantial headway has been made in characterizing C9orf72-mediated disease and unravelling its underlying aetiopathogenesis. Three main disease mechanisms have been proposed: loss of function of the C9orf72 protein and toxic gain of function from C9orf72 repeat RNA or from dipeptide repeat proteins produced by repeat-associated non-ATG translation. Several downstream processes across a range of cellular functions have also been implicated. In this article, we review the pathological and mechanistic features of C9orf72-associated FTD and ALS (collectively termed C9FTD/ALS), the model systems used to study these conditions, and the probable initiators of downstream disease mechanisms. We suggest that a combination of upstream mechanisms involving both loss and gain of function and downstream cellular pathways involving both cell-autonomous and non-cell-autonomous effects contributes to disease progression.
Collapse
Affiliation(s)
- Rubika Balendra
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, UCL, London, UK
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK. .,UK Dementia Research Institute at UCL, UCL Institute of Neurology, London, UK.
| |
Collapse
|
128
|
Solomon DA, Stepto A, Au WH, Adachi Y, Diaper DC, Hall R, Rekhi A, Boudi A, Tziortzouda P, Lee YB, Smith B, Bridi JC, Spinelli G, Dearlove J, Humphrey DM, Gallo JM, Troakes C, Fanto M, Soller M, Rogelj B, Parsons RB, Shaw CE, Hortobágyi T, Hirth F. A feedback loop between dipeptide-repeat protein, TDP-43 and karyopherin-α mediates C9orf72-related neurodegeneration. Brain 2019; 141:2908-2924. [PMID: 30239641 PMCID: PMC6158706 DOI: 10.1093/brain/awy241] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022] Open
Abstract
Accumulation and aggregation of TDP-43 is a major pathological hallmark of amyotrophic lateral sclerosis and frontotemporal dementia. TDP-43 inclusions also characterize patients with GGGGCC (G4C2) hexanucleotide repeat expansion in C9orf72 that causes the most common genetic form of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD). Functional studies in cell and animal models have identified pathogenic mechanisms including repeat-induced RNA toxicity and accumulation of G4C2-derived dipeptide-repeat proteins. The role of TDP-43 dysfunction in C9ALS/FTD, however, remains elusive. We found G4C2-derived dipeptide-repeat protein but not G4C2-RNA accumulation caused TDP-43 proteinopathy that triggered onset and progression of disease in Drosophila models of C9ALS/FTD. Timing and extent of TDP-43 dysfunction was dependent on levels and identity of dipeptide-repeat proteins produced, with poly-GR causing early and poly-GA/poly-GP causing late onset of disease. Accumulating cytosolic, but not insoluble aggregated TDP-43 caused karyopherin-α2/4 (KPNA2/4) pathology, increased levels of dipeptide-repeat proteins and enhanced G4C2-related toxicity. Comparable KPNA4 pathology was observed in both sporadic frontotemporal dementia and C9ALS/FTD patient brains characterized by its nuclear depletion and cytosolic accumulation, irrespective of TDP-43 or dipeptide-repeat protein aggregates. These findings identify a vicious feedback cycle for dipeptide-repeat protein-mediated TDP-43 and subsequent KPNA pathology, which becomes self-sufficient of the initiating trigger and causes C9-related neurodegeneration.
Collapse
Affiliation(s)
- Daniel A Solomon
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Alan Stepto
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Wing Hei Au
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Yoshitsugu Adachi
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Danielle C Diaper
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Rachel Hall
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Anjeet Rekhi
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Adel Boudi
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Paraskevi Tziortzouda
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Youn-Bok Lee
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Bradley Smith
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Jessika C Bridi
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Greta Spinelli
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Jonah Dearlove
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Dickon M Humphrey
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Jean-Marc Gallo
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Claire Troakes
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Manolis Fanto
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Matthias Soller
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Boris Rogelj
- Jozef Stefan Institute, Department of Biotechnology and Biomedical Research Institute BRIS and University of Ljubljana, Faculty of Chemistry and Chemical Technology, Ljubljana, Slovenia
| | - Richard B Parsons
- King's College London, School of Cancer Studies and Pharmaceutical Sciences, London, UK
| | - Christopher E Shaw
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Tibor Hortobágyi
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, Departments of Neurology and Neuropathology, University of Debrecen, Debrecen, Hungary.,King's College London, Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Frank Hirth
- King's College London, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| |
Collapse
|
129
|
Banez-Coronel M, Ranum LPW. Repeat-associated non-AUG (RAN) translation: insights from pathology. J Transl Med 2019; 99:929-942. [PMID: 30918326 PMCID: PMC7219275 DOI: 10.1038/s41374-019-0241-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
More than 40 different neurological diseases are caused by microsatellite repeat expansions. Since the discovery of repeat-associated non-AUG (RAN) translation by Zu et al. in 2011, nine expansion disorders have been identified as RAN-positive diseases. RAN proteins are translated from different types of nucleotide repeat expansions and can be produced from both sense and antisense transcripts. In some diseases, RAN proteins have been shown to accumulate in affected brain regions. Here we review the pathological and molecular aspects associated with RAN protein accumulation for each particular disorder, the correlation between disease pathology and the available in vivo models and the common aspects shared by some of the newly discovered RAN proteins.
Collapse
Affiliation(s)
- Monica Banez-Coronel
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, 32610, USA
| | - Laura P W Ranum
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA.
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
- Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
130
|
Kearse MG, Goldman DH, Choi J, Nwaezeapu C, Liang D, Green KM, Goldstrohm AC, Todd PK, Green R, Wilusz JE. Ribosome queuing enables non-AUG translation to be resistant to multiple protein synthesis inhibitors. Genes Dev 2019; 33:871-885. [PMID: 31171704 PMCID: PMC6601509 DOI: 10.1101/gad.324715.119] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/24/2019] [Indexed: 02/05/2023]
Abstract
Aberrant translation initiation at non-AUG start codons is associated with multiple cancers and neurodegenerative diseases. Nevertheless, how non-AUG translation may be regulated differently from canonical translation is poorly understood. Here, we used start codon-specific reporters and ribosome profiling to characterize how translation from non-AUG start codons responds to protein synthesis inhibitors in human cells. These analyses surprisingly revealed that translation of multiple non-AUG-encoded reporters and the endogenous GUG-encoded DAP5 (eIF4G2/p97) mRNA is resistant to cycloheximide (CHX), a translation inhibitor that severely slows but does not completely abrogate elongation. Our data suggest that slowly elongating ribosomes can lead to queuing/stacking of scanning preinitiation complexes (PICs), preferentially enhancing recognition of weak non-AUG start codons. Consistent with this model, limiting PIC formation or scanning sensitizes non-AUG translation to CHX. We further found that non-AUG translation is resistant to other inhibitors that target ribosomes within the coding sequence but not those targeting newly initiated ribosomes. Together, these data indicate that ribosome queuing enables mRNAs with poor initiation context-namely, those with non-AUG start codons-to be resistant to pharmacological translation inhibitors at concentrations that robustly inhibit global translation.
Collapse
Affiliation(s)
- Michael G Kearse
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Daniel H Goldman
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Jiou Choi
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Chike Nwaezeapu
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Dongming Liang
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Katelyn M Green
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Aaron C Goldstrohm
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Peter K Todd
- Department of Neurology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Veterans Affairs Medical Center, Ann Arbor, Michigan 48105, USA
| | - Rachel Green
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Jeremy E Wilusz
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
131
|
Rodriguez CM, Todd PK. New pathologic mechanisms in nucleotide repeat expansion disorders. Neurobiol Dis 2019; 130:104515. [PMID: 31229686 DOI: 10.1016/j.nbd.2019.104515] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/07/2019] [Accepted: 06/19/2019] [Indexed: 12/14/2022] Open
Abstract
Tandem microsatellite repeats are common throughout the human genome and intrinsically unstable, exhibiting expansions and contractions both somatically and across generations. Instability in a small subset of these repeats are currently linked to human disease, although recent findings suggest more disease-causing repeats await discovery. These nucleotide repeat expansion disorders (NREDs) primarily affect the nervous system and commonly lead to neurodegeneration through toxic protein gain-of-function, protein loss-of-function, and toxic RNA gain-of-function mechanisms. However, the lines between these categories have blurred with recent findings of unconventional Repeat Associated Non-AUG (RAN) translation from putatively non-coding regions of the genome. Here we review two emerging topics in NREDs: 1) The mechanisms by which RAN translation occurs and its role in disease pathogenesis and 2) How nucleotide repeats as RNA and translated proteins influence liquid-liquid phase separation, membraneless organelle dynamics, and nucleocytoplasmic transport. We examine these topics with a particular eye on two repeats: the CGG repeat expansion responsible for Fragile X syndrome and Fragile X-associated Tremor Ataxia Syndrome (FXTAS) and the intronic GGGGCC repeat expansion in C9orf72, the most common inherited cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Our thesis is that these emerging disease mechanisms can inform a broader understanding of the native roles of microsatellites in cellular function and that aberrations in these native processes provide clues to novel therapeutic strategies for these currently untreatable disorders.
Collapse
Affiliation(s)
- C M Rodriguez
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | - P K Todd
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA; VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| |
Collapse
|
132
|
Xu W, Bao P, Jiang X, Wang H, Qin M, Wang R, Wang T, Yang Y, Lorenzini I, Liao L, Sattler R, Xu J. Reactivation of nonsense-mediated mRNA decay protects against C9orf72 dipeptide-repeat neurotoxicity. Brain 2019; 142:1349-1364. [PMID: 30938419 PMCID: PMC6487333 DOI: 10.1093/brain/awz070] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/04/2019] [Accepted: 01/27/2019] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis is a deleterious neurodegenerative disease without effective treatment options. Recent studies have indicated the involvement of the dysregulation of RNA metabolism in the pathogenesis of amyotrophic lateral sclerosis. Among the various RNA regulatory machineries, nonsense-mediated mRNA decay (NMD) is a stress responsive cellular surveillance system that degrades selected mRNA substrates to prevent the translation of defective or harmful proteins. Whether this pathway is affected in neurodegenerative diseases is unclear. Here we report the inhibition of NMD by arginine-rich dipeptide repeats derived from C9orf72 hexanucleotide repeat expansion, the most common cause of familial amyotrophic lateral sclerosis. Bioinformatic analysis of multiple transcriptome profiles revealed significant overlap of upregulated genes in NMD-defective cells with those in the brain tissues, micro-dissected motor neurons, or induced pluripotent stem cell-derived motor neurons specifically from amyotrophic lateral sclerosis patients carrying C9orf72 hexanucleotide repeat expansion, suggesting the suppression of NMD pathway in these patients. Using Drosophila as a model, we have validated that the C9orf72 hexanucleotide repeat expansion products could lead to the accumulation of the NMD substrates and identified arginine-rich dipeptide repeats, including poly glycine-arginine and poly proline-arginine, as the main culprits of NMD inhibition. Furthermore, in human SH-SY5Y neuroblastoma cells and in mouse brains, expression of glycine-arginine with 36 repeats (GR36) was sufficient to cause NMD inhibition. In cells expressing GR36, stress granule accumulation was accompanied by decreased processing body formation, which contributed to the inhibition of NMD. Remarkably, expression of UPF1, a core gene in the NMD pathway, efficiently blocked neurotoxicity caused by arginine-rich dipeptide repeats in both cellular and Drosophila models. Although not as effective as UPF1, expression of another NMD gene UPF2 also ameliorated the degenerative phenotypes in dipeptide repeat-expressing flies, indicating that genetically reactivating the NMD pathway could suppress dipeptide repeat toxicity. Finally, after validating tranilast as an NMD-activating drug, we demonstrated the therapeutic potential of this asthma drug in cellular and Drosophila models of C9orf72 dipeptide repeat neurotoxicity. Therefore, our study has revealed a cellular mechanism whereby arginine-rich C9orf72 dipeptide repeats could inhibit NMD activities by reducing the abundance of processing bodies. Furthermore, our results suggested that activation of the NMD pathway could be a potential therapeutic strategy for amyotrophic lateral sclerosis with defective RNA metabolism.
Collapse
Affiliation(s)
- Wangchao Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Puhua Bao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xin Jiang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Haifang Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Meiling Qin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ruiqi Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tao Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Shanghai, China
| | - Ileana Lorenzini
- Barrow Neurological Institute, Dignity Health, St. Joseph’s Hospital and Medical Center, Phoenix AZ, USA
| | - Lujian Liao
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Rita Sattler
- Barrow Neurological Institute, Dignity Health, St. Joseph’s Hospital and Medical Center, Phoenix AZ, USA
| | - Jin Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
133
|
Goodman LD, Prudencio M, Srinivasan AR, Rifai OM, Lee VMY, Petrucelli L, Bonini NM. eIF4B and eIF4H mediate GR production from expanded G4C2 in a Drosophila model for C9orf72-associated ALS. Acta Neuropathol Commun 2019; 7:62. [PMID: 31023341 PMCID: PMC6485101 DOI: 10.1186/s40478-019-0711-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/25/2019] [Indexed: 01/09/2023] Open
Abstract
The discovery of an expanded (GGGGCC)n repeat (termed G4C2) within the first intron of C9orf72 in familial ALS/FTD has led to a number of studies showing that the aberrant expression of G4C2 RNA can produce toxic dipeptides through repeat-associated non-AUG (RAN-) translation. To reveal canonical translation factors that impact this process, an unbiased loss-of-function screen was performed in a G4C2 fly model that maintained the upstream intronic sequence of the human gene and contained a GFP tag in the GR reading frame. 11 of 48 translation factors were identified that impact production of the GR-GFP protein. Further investigations into two of these, eIF4B and eIF4H, revealed that downregulation of these factors reduced toxicity caused by the expression of expanded G4C2 and reduced production of toxic GR dipeptides from G4C2 transcripts. In patient-derived cells and in post-mortem tissue from ALS/FTD patients, eIF4H was found to be downregulated in cases harboring the G4C2 mutation compared to patients lacking the mutation and healthy individuals. Overall, these data define eIF4B and eIF4H as disease modifiers whose activity is important for RAN-translation of the GR peptide from G4C2-transcripts.
Collapse
Affiliation(s)
- Lindsey D. Goodman
- 0000 0004 1936 8972grid.25879.31Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Mercedes Prudencio
- 0000 0004 0443 9942grid.417467.7Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Ananth R. Srinivasan
- 0000 0004 1936 8972grid.25879.31Department of Biology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Olivia M. Rifai
- 0000 0004 1936 8972grid.25879.31Department of Biology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Virginia M.-Y. Lee
- 0000 0004 1936 8972grid.25879.31Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Leonard Petrucelli
- 0000 0004 0443 9942grid.417467.7Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Nancy M. Bonini
- 0000 0004 1936 8972grid.25879.31Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA ,0000 0004 1936 8972grid.25879.31Department of Biology, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
134
|
White MR, Mitrea DM, Zhang P, Stanley CB, Cassidy DE, Nourse A, Phillips AH, Tolbert M, Taylor JP, Kriwacki RW. C9orf72 Poly(PR) Dipeptide Repeats Disturb Biomolecular Phase Separation and Disrupt Nucleolar Function. Mol Cell 2019; 74:713-728.e6. [PMID: 30981631 DOI: 10.1016/j.molcel.2019.03.019] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/12/2019] [Accepted: 03/18/2019] [Indexed: 12/11/2022]
Abstract
Repeat expansion in the C9orf72 gene is the most common cause of the neurodegenerative disorder amyotrophic lateral sclerosis (C9-ALS) and is linked to the unconventional translation of five dipeptide-repeat polypeptides (DPRs). The two enriched in arginine, poly(GR) and poly(PR), infiltrate liquid-like nucleoli, co-localize with the nucleolar protein nucleophosmin (NPM1), and alter the phase separation behavior of NPM1 in vitro. Here, we show that poly(PR) DPRs bind tightly to a long acidic tract within the intrinsically disordered region of NPM1, altering its phase separation with nucleolar partners to the extreme of forming large, soluble complexes that cause droplet dissolution in vitro. In cells, poly(PR) DPRs disperse NPM1 from nucleoli and entrap rRNA in static condensates in a DPR-length-dependent manner. We propose that R-rich DPR toxicity involves disrupting the role of phase separation by NPM1 in organizing ribosomal proteins and RNAs within the nucleolus.
Collapse
Affiliation(s)
- Michael R White
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Diana M Mitrea
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Peipei Zhang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christopher B Stanley
- Large Scale Structures Group, Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Devon E Cassidy
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Amanda Nourse
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Molecular Interaction Analysis Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Aaron H Phillips
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michele Tolbert
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - J Paul Taylor
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Howard Hughes Medical Institute, Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richard W Kriwacki
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Sciences Center, Memphis, TN 38105, USA.
| |
Collapse
|
135
|
Delaidelli A, Jan A, Herms J, Sorensen PH. Translational control in brain pathologies: biological significance and therapeutic opportunities. Acta Neuropathol 2019; 137:535-555. [PMID: 30739199 DOI: 10.1007/s00401-019-01971-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
Abstract
Messenger RNA (mRNA) translation is the terminal step in protein synthesis, providing a crucial regulatory checkpoint for this process. Translational control allows specific cell types to respond to rapid changes in the microenvironment or to serve specific functions. For example, neurons use mRNA transport to achieve local protein synthesis at significant distances from the nucleus, the site of RNA transcription. Altered expression or functions of the various components of the translational machinery have been linked to several pathologies in the central nervous system. In this review, we provide a brief overview of the basic principles of mRNA translation, and discuss alterations of this process relevant to CNS disease conditions, with a focus on brain tumors and chronic neurological conditions. Finally, synthesizing this knowledge, we discuss the opportunities to exploit the biology of altered mRNA translation for novel therapies in brain disorders, as well as how studying these alterations can shed new light on disease mechanisms.
Collapse
Affiliation(s)
- Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | - Asad Jan
- Department of Biomedicine, Aarhus Institute of Advanced Studies, Aarhus University, Høegh-Guldbergs Gade 6B, 8000, Aarhus C, Denmark
| | - Jochen Herms
- Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilians-University Munich, Schillerstraße 44, 80336, Munich, Germany
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
136
|
Nguyen L, Cleary JD, Ranum LPW. Repeat-Associated Non-ATG Translation: Molecular Mechanisms and Contribution to Neurological Disease. Annu Rev Neurosci 2019; 42:227-247. [PMID: 30909783 DOI: 10.1146/annurev-neuro-070918-050405] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Microsatellite mutations involving the expansion of tri-, tetra-, penta-, or hexanucleotide repeats cause more than 40 different neurological disorders. Although, traditionally, the position of the repeat within or outside of an open reading frame has been used to focus research on disease mechanisms involving protein loss of function, protein gain of function, or RNA gain of function, the discoveries of bidirectional transcription and repeat-associated non-ATG (RAN) have blurred these distinctions. Here we review what is known about RAN proteins in disease, the mechanisms by which they are produced, and the novel therapeutic opportunities they provide.
Collapse
Affiliation(s)
- Lien Nguyen
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA;
| | - John Douglas Cleary
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA;
| | - Laura P W Ranum
- Center for NeuroGenetics, Department of Molecular Genetics and Microbiology, Genetics Institute, and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA;
| |
Collapse
|
137
|
Chew J, Cook C, Gendron TF, Jansen-West K, Del Rosso G, Daughrity LM, Castanedes-Casey M, Kurti A, Stankowski JN, Disney MD, Rothstein JD, Dickson DW, Fryer JD, Zhang YJ, Petrucelli L. Aberrant deposition of stress granule-resident proteins linked to C9orf72-associated TDP-43 proteinopathy. Mol Neurodegener 2019; 14:9. [PMID: 30767771 PMCID: PMC6377782 DOI: 10.1186/s13024-019-0310-z] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/08/2019] [Indexed: 12/14/2022] Open
Abstract
Background A G4C2 hexanucleotide repeat expansion in the noncoding region of C9orf72 is the major genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis (c9FTD/ALS). Putative disease mechanisms underlying c9FTD/ALS include toxicity from sense G4C2 and antisense G2C4 repeat-containing RNA, and from dipeptide repeat (DPR) proteins unconventionally translated from these RNA products. Methods Intracerebroventricular injections with adeno-associated virus (AAV) encoding 2 or 149 G4C2 repeats were performed on postnatal day 0, followed by assessment of behavioral and neuropathological phenotypes. Results Relative to control mice, gliosis and neurodegeneration accompanied by cognitive and motor deficits were observed in (G4C2)149 mice by 6 months of age. Recapitulating key pathological hallmarks, we also demonstrate that sense and antisense RNA foci, inclusions of poly(GA), poly(GP), poly(GR), poly(PR), and poly(PA) DPR proteins, and inclusions of endogenous phosphorylated TDP-43 (pTDP-43) developed in (G4C2)149 mice but not control (G4C2)2 mice. Notably, proteins that play a role in the regulation of stress granules – RNA-protein assemblies that form in response to translational inhibition and that have been implicated in c9FTD/ALS pathogenesis – were mislocalized in (G4C2)149 mice as early as 3 months of age. Specifically, we observed the abnormal deposition of stress granule components within inclusions immunopositive for poly(GR) and pTDP-43, as well as evidence of nucleocytoplasmic transport defects. Conclusions Our in vivo model of c9FTD/ALS is the first to robustly recapitulate hallmark features derived from both sense and antisense C9orf72 repeat-associated transcripts complete with neurodegeneration and behavioral impairments. More importantly, the early appearance of persistent pathological stress granules prior to significant pTDP-43 deposition implicates an aberrant stress granule response as a key disease mechanism driving TDP-43 proteinopathy in c9FTD/ALS. Electronic supplementary material The online version of this article (10.1186/s13024-019-0310-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jeannie Chew
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Casey Cook
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.,Neurobiology of Disease Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, 4500 San Pablo Rd, Jacksonville, Florida, 32224, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.,Neurobiology of Disease Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, 4500 San Pablo Rd, Jacksonville, Florida, 32224, USA
| | - Karen Jansen-West
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Giulia Del Rosso
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Lillian M Daughrity
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Monica Castanedes-Casey
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Jeannette N Stankowski
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Matthew D Disney
- Department of Chemistry, The Scripps Research Institute, Scripps Florida, 130 Scripps Way #3A1, Jupiter, Florida, 33458, USA
| | - Jeffrey D Rothstein
- Department of Neurology, Brain Science Institute, Johns Hopkins University, 855 N Wolfe St, Baltimore, MD, 21205, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.,Neurobiology of Disease Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, 4500 San Pablo Rd, Jacksonville, Florida, 32224, USA
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.,Neurobiology of Disease Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, 4500 San Pablo Rd, Jacksonville, Florida, 32224, USA
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA. .,Neurobiology of Disease Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, 4500 San Pablo Rd, Jacksonville, Florida, 32224, USA.
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic College of Medicine, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA. .,Neurobiology of Disease Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, 4500 San Pablo Rd, Jacksonville, Florida, 32224, USA.
| |
Collapse
|
138
|
Westergard T, McAvoy K, Russell K, Wen X, Pang Y, Morris B, Pasinelli P, Trotti D, Haeusler A. Repeat-associated non-AUG translation in C9orf72-ALS/FTD is driven by neuronal excitation and stress. EMBO Mol Med 2019; 11:e9423. [PMID: 30617154 PMCID: PMC6365928 DOI: 10.15252/emmm.201809423] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/06/2018] [Indexed: 01/07/2023] Open
Abstract
Nucleotide repeat expansions (NREs) are prevalent mutations in a multitude of neurodegenerative diseases. Repeat-associated non-AUG (RAN) translation of these repeat regions produces mono or dipeptides that contribute to the pathogenesis of these diseases. However, the mechanisms and drivers of RAN translation are not well understood. Here we analyzed whether different cellular stressors promote RAN translation of dipeptide repeats (DPRs) associated with the G4C2 hexanucleotide expansions in C9orf72, the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). We found that activating glutamate receptors or optogenetically increasing neuronal activity by repetitive trains of depolarization induced DPR formation in primary cortical neurons and patient derived spinal motor neurons. Increases in the integrated stress response (ISR) were concomitant with increased RAN translation of DPRs, both in neurons and different cell lines. Targeting phosphorylated-PERK and the phosphorylated-eif2α complex reduces DPR levels revealing a potential therapeutic strategy to attenuate DPR-dependent disease pathogenesis in NRE-linked diseases.
Collapse
Affiliation(s)
- Thomas Westergard
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson University, Philadelphia, PA, USA
| | - Kevin McAvoy
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson University, Philadelphia, PA, USA
| | - Katelyn Russell
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson University, Philadelphia, PA, USA
| | - Xinmei Wen
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson University, Philadelphia, PA, USA
| | - Yu Pang
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson University, Philadelphia, PA, USA
| | - Brandie Morris
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson University, Philadelphia, PA, USA
| | - Piera Pasinelli
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson University, Philadelphia, PA, USA
| | - Davide Trotti
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson University, Philadelphia, PA, USA
| | - Aaron Haeusler
- Department of Neuroscience, Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
139
|
Weskamp K, Safren N, Miguez R, Barmada S. Monitoring Neuronal Survival via Longitudinal Fluorescence Microscopy. J Vis Exp 2019. [PMID: 30735193 DOI: 10.3791/59036] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Standard cytotoxicity assays, which require the collection of lysates or fixed cells at multiple time points, have limited sensitivity and capacity to assess factors that influence neuronal fate. These assays require the observation of separate populations of cells at discrete time points. As a result, individual cells cannot be followed prospectively over time, severely limiting the ability to discriminate whether subcellular events, such as puncta formation or protein mislocalization, are pathogenic drivers of disease, homeostatic responses, or merely coincidental phenomena. Single-cell longitudinal microscopy overcomes these limitations, allowing the researcher to determine differences in survival between populations and draw causal relationships with enhanced sensitivity. This video guide will outline a representative workflow for experiments measuring single-cell survival of rat primary cortical neurons expressing a fluorescent protein marker. The viewer will learn how to achieve high-efficiency transfections, collect and process images enabling the prospective tracking of individual cells, and compare the relative survival of neuronal populations using Cox proportional hazards analysis.
Collapse
Affiliation(s)
- Kaitlin Weskamp
- Department of Neurology, University of Michigan School of Medicine; Neuroscience Graduate Program, University of Michigan School of Medicine
| | - Nathaniel Safren
- Department of Neurology, University of Michigan School of Medicine
| | - Roberto Miguez
- Department of Neurology, University of Michigan School of Medicine
| | - Sami Barmada
- Department of Neurology, University of Michigan School of Medicine; Neuroscience Graduate Program, University of Michigan School of Medicine;
| |
Collapse
|
140
|
Molecular Mechanisms of Neurodegeneration Related to C9orf72 Hexanucleotide Repeat Expansion. Behav Neurol 2019; 2019:2909168. [PMID: 30774737 PMCID: PMC6350563 DOI: 10.1155/2019/2909168] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/28/2018] [Accepted: 09/18/2018] [Indexed: 12/11/2022] Open
Abstract
Two clinically distinct diseases, amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), have recently been classified as two extremes of the FTD/ALS spectrum. The neuropathological correlate of FTD is frontotemporal lobar degeneration (FTLD), characterized by tau-, TDP-43-, and FUS-immunoreactive neuronal inclusions. An earlier discovery that a hexanucleotide repeat expansion mutation in chromosome 9 open reading frame 72 (C9orf72) gene causes ALS and FTD established a special subtype of ALS and FTLD with TDP-43 pathology (C9FTD/ALS). Normal individuals carry 2–10 hexanucleotide GGGGCC repeats in the C9orf72 gene, while more than a few hundred repeats represent a risk for ALS and FTD. The proposed molecular mechanisms by which C9orf72 repeat expansions induce neurodegenerative changes are C9orf72 loss-of-function through haploinsufficiency, RNA toxic gain-of-function, and gain-of-function through the accumulation of toxic dipeptide repeat proteins. However, many more cellular processes are affected by pathological processes in C9FTD/ALS, including nucleocytoplasmic transport, RNA processing, normal function of nucleolus, formation of membraneless organelles, translation, ubiquitin proteasome system, Notch signalling pathway, granule transport, and normal function of TAR DNA-binding protein 43 (TDP-43). Although the exact molecular mechanisms through which C9orf72 repeat expansions account for neurodegeneration have not been elucidated, some potential therapeutics, such as antisense oligonucleotides targeting hexanucleotide GGGGCC repeats in mRNA, were successful in preclinical trials and are awaiting phase 1 clinical trials. In this review, we critically discuss each proposed mechanism and provide insight into the most recent studies aiming to elucidate the molecular underpinnings of C9FTD/ALS.
Collapse
|
141
|
Abstract
Microsatellite expansions cause more than 40 neurological disorders, including Huntington's disease, myotonic dystrophy, and C9ORF72 amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD). These repeat expansion mutations can produce repeat-associated non-ATG (RAN) proteins in all three reading frames, which accumulate in disease-relevant tissues. There has been considerable interest in RAN protein products and their downstream consequences, particularly for the dipeptide proteins found in C9ORF72 ALS/FTD. Understanding how RAN translation occurs, what cellular factors contribute to RAN protein accumulation, and how these proteins contribute to disease should lead to a better understanding of the basic mechanisms of gene expression and human disease.
Collapse
Affiliation(s)
- John Douglas Cleary
- From the Center for NeuroGenetics
- Departments of Molecular Genetics and Microbiology and
- Genetics Institute, and
| | - Amrutha Pattamatta
- From the Center for NeuroGenetics
- Departments of Molecular Genetics and Microbiology and
- Genetics Institute, and
| | - Laura P W Ranum
- From the Center for NeuroGenetics,
- Departments of Molecular Genetics and Microbiology and
- Genetics Institute, and
- Neurology, College of Medicine
- McKnight Brain Institute, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
142
|
Glineburg MR, Todd PK, Charlet-Berguerand N, Sellier C. Repeat-associated non-AUG (RAN) translation and other molecular mechanisms in Fragile X Tremor Ataxia Syndrome. Brain Res 2018; 1693:43-54. [PMID: 29453961 PMCID: PMC6010627 DOI: 10.1016/j.brainres.2018.02.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/31/2018] [Accepted: 02/02/2018] [Indexed: 11/11/2022]
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset inherited neurodegenerative disorder characterized by progressive intention tremor, gait ataxia and dementia associated with mild brain atrophy. The cause of FXTAS is a premutation expansion, of 55 to 200 CGG repeats localized within the 5'UTR of FMR1. These repeats are transcribed in the sense and antisense directions into mutants RNAs, which have increased expression in FXTAS. Furthermore, CGG sense and CCG antisense expanded repeats are translated into novel proteins despite their localization in putatively non-coding regions of the transcript. Here we focus on two proposed disease mechanisms for FXTAS: 1) RNA gain-of-function, whereby the mutant RNAs bind specific proteins and preclude their normal functions, and 2) repeat-associated non-AUG (RAN) translation, whereby translation through the CGG or CCG repeats leads to the production of toxic homopolypeptides, which in turn interfere with a variety of cellular functions. Here, we analyze the data generated to date on both of these potential molecular mechanisms and lay out a path forward for determining which factors drive FXTAS pathogenicity.
Collapse
Affiliation(s)
| | - Peter K Todd
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; Veteran's Affairs Medical Center, Ann Arbor, MI 48105, USA
| | - Nicolas Charlet-Berguerand
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, University of Strasbourg, 67400 Illkirch, France
| | - Chantal Sellier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, University of Strasbourg, 67400 Illkirch, France.
| |
Collapse
|
143
|
Richards RI, Robertson SA, Kastner DL. Neurodegenerative diseases have genetic hallmarks of autoinflammatory disease. Hum Mol Genet 2018; 27:R108-R118. [PMID: 29684205 PMCID: PMC6061832 DOI: 10.1093/hmg/ddy139] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 03/12/2018] [Accepted: 04/16/2018] [Indexed: 12/25/2022] Open
Abstract
The notion that one common pathogenic pathway could account for the various clinically distinguishable, typically late-onset neurodegenerative diseases might appear unlikely given the plethora of diverse primary causes of neurodegeneration. On the contrary, an autoinflammatory pathogenic mechanism allows diverse genetic and environmental factors to converge into a common chain of causality. Inflammation has long been known to correlate with neurodegeneration. Until recently this relationship was seen as one of consequence rather than cause-with inflammatory cells and events acting to 'clean up the mess' after neurological injury. This explanation is demonstrably inadequate and it is now clear that inflammation is at the very least, rate-limiting for neurodegeneration (and more likely, a principal underlying cause in most if not all neurodegenerative diseases), protective in its initial acute phase, but pernicious in its latter chronic phase.
Collapse
Affiliation(s)
- Robert I Richards
- Department of Molecular and Biomedical Sciences, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Sarah A Robertson
- Robinson Research Institute, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Daniel L Kastner
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
144
|
|
145
|
Rudich P, Lamitina T. Models and mechanisms of repeat expansion disorders: a worm's eye view. J Genet 2018; 97:665-677. [PMID: 30027902 PMCID: PMC6482835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The inappropriate genetic expansion of various repetitive DNA sequences underlies over 20 distinct inherited diseases. The genetic context of these repeats in exons, introns and untranslated regions has played a major role in thinking about the mechanisms by which various repeat expansions might cause disease. Repeat expansions in exons are thought to give rise to expanded toxic protein repeats (i.e. polyQ). Repeat expansions in introns and UTRs (i.e. FXTAS) are thought to produce aberrant repeat-bearing RNAs that interact with and sequester a wide variety of essential proteins, resulting in cellular toxicity. However, a new phenomenon termed 'repeat-associated nonAUG dependent (RAN) translation' paints a new and unifying picture of how distinct repeat expansion-bearing RNAs might act as substrates for this noncanonical form of translation, leading to the production of a wide range of repeat sequence-specific-encoded toxic proteins. Here, we review how the model system Caenorhabditis elegans has been utilized to model many repeat disorders and discuss how RAN translation could be a previously unappreciated contributor to the toxicity associated with these different models.
Collapse
Affiliation(s)
- Paige Rudich
- Graduate Program in Cell Biology and Molecular Physiology, University of Pittsburgh Medical Center, Pittsburgh, PA 15224, USA.
| | | |
Collapse
|
146
|
Poly(GR) impairs protein translation and stress granule dynamics in C9orf72-associated frontotemporal dementia and amyotrophic lateral sclerosis. Nat Med 2018; 24:1136-1142. [PMID: 29942091 DOI: 10.1038/s41591-018-0071-1] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/30/2018] [Indexed: 12/13/2022]
Abstract
The major genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) is a C9orf72 G4C2 repeat expansion1,2. Proposed mechanisms by which the expansion causes c9FTD/ALS include toxicity from repeat-containing RNA and from dipeptide repeat proteins translated from these transcripts. To investigate the contribution of poly(GR) dipeptide repeat proteins to c9FTD/ALS pathogenesis in a mammalian in vivo model, we generated mice that expressed GFP-(GR)100 in the brain. GFP-(GR)100 mice developed age-dependent neurodegeneration, brain atrophy, and motor and memory deficits through the accumulation of diffuse, cytoplasmic poly(GR). Poly(GR) co-localized with ribosomal subunits and the translation initiation factor eIF3η in GFP-(GR)100 mice and, of importance, in c9FTD/ALS patients. Combined with the differential expression of ribosome-associated genes in GFP-(GR)100 mice, these findings demonstrate poly(GR)-mediated ribosomal distress. Indeed, poly(GR) inhibited canonical and non-canonical protein translation in HEK293T cells, and also induced the formation of stress granules and delayed their disassembly. These data suggest that poly(GR) contributes to c9FTD/ALS by impairing protein translation and stress granule dynamics, consequently causing chronic cellular stress and preventing cells from mounting an effective stress response. Decreasing poly(GR) and/or interrupting interactions between poly(GR) and ribosomal and stress granule-associated proteins may thus represent potential therapeutic strategies to restore homeostasis.
Collapse
|
147
|
Sonobe Y, Ghadge G, Masaki K, Sendoel A, Fuchs E, Roos RP. Translation of dipeptide repeat proteins from the C9ORF72 expanded repeat is associated with cellular stress. Neurobiol Dis 2018; 116:155-165. [PMID: 29792928 DOI: 10.1016/j.nbd.2018.05.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 05/17/2018] [Accepted: 05/21/2018] [Indexed: 01/13/2023] Open
Abstract
Expansion of a hexanucleotide repeat (HRE), GGGGCC, in the C9ORF72 gene is recognized as the most common cause of familial amyotrophic lateral sclerosis (FALS), frontotemporal dementia (FTD) and ALS-FTD, as well as 5-10% of sporadic ALS. Despite the location of the HRE in the non-coding region (with respect to the main C9ORF72 gene product), dipeptide repeat proteins (DPRs) that are thought to be toxic are translated from the HRE in all three reading frames from both the sense and antisense transcript. Here, we identified a CUG that has a good Kozak consensus sequence as the translation initiation codon. Mutation of this CTG significantly suppressed polyglycine-alanine (GA) translation. GA was translated when the G4C2 construct was placed as the second cistron in a bicistronic construct. CRISPR/Cas9-induced knockout of a non-canonical translation initiation factor, eIF2A, impaired GA translation. Transfection of G4C2 constructs induced an integrated stress response (ISR), while triggering the ISR led to a continuation of translation of GA with a decline in conventional cap-dependent translation. These in vitro observations were confirmed in chick embryo neural cells. The findings suggest that DPRs translated from an HRE in C9ORF72 aggregate and lead to an ISR that then leads to continuing DPR production and aggregation, thereby creating a continuing pathogenic cycle.
Collapse
Affiliation(s)
- Yoshifumi Sonobe
- Department of Neurology, University of Chicago Medical Center, 5841 S. Maryland Ave., Chicago, IL 60637, United States
| | - Ghanashyam Ghadge
- Department of Neurology, University of Chicago Medical Center, 5841 S. Maryland Ave., Chicago, IL 60637, United States
| | - Katsuhisa Masaki
- Department of Neurology, University of Chicago Medical Center, 5841 S. Maryland Ave., Chicago, IL 60637, United States
| | - Ataman Sendoel
- Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Ave., Box 300, NY, NY, 10021-6399, United States
| | - Elaine Fuchs
- Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Ave., Box 300, NY, NY, 10021-6399, United States
| | - Raymond P Roos
- Department of Neurology, University of Chicago Medical Center, 5841 S. Maryland Ave., Chicago, IL 60637, United States.
| |
Collapse
|
148
|
Moon SL, Sonenberg N, Parker R. Neuronal Regulation of eIF2α Function in Health and Neurological Disorders. Trends Mol Med 2018; 24:575-589. [PMID: 29716790 DOI: 10.1016/j.molmed.2018.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 12/12/2022]
Abstract
A key site of translation control is the phosphorylation of the eukaryotic translation initiation factor 2α (eIF2α), which reduces the rate of GDP to GTP exchange by eIF2B, leading to altered translation. The extent of eIF2α phosphorylation within neurons can alter synaptic plasticity. Phosphorylation of eIF2α is triggered by four stress-responsive kinases, and as such eIF2α is often phosphorylated during neurological perturbations or disease. Moreover, in some cases decreasing eIF2α phosphorylation mitigates neurodegeneration, suggesting that this could be a therapeutic target. Mutations in the γ subunit of eIF2, the guanine exchange factor eIF2B, an eIF2α phosphatase, or in two eIF2α kinases can cause disease in humans, demonstrating the importance of proper regulation of eIF2α phosphorylation for health.
Collapse
Affiliation(s)
- Stephanie L Moon
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Roy Parker
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80303, USA; Howard Hughes Medical Institute, University of Colorado, Boulder, CO 80303, USA.
| |
Collapse
|
149
|
Shirokikh NE, Preiss T. Translation initiation by cap-dependent ribosome recruitment: Recent insights and open questions. WILEY INTERDISCIPLINARY REVIEWS-RNA 2018; 9:e1473. [PMID: 29624880 DOI: 10.1002/wrna.1473] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/02/2018] [Accepted: 02/14/2018] [Indexed: 12/14/2022]
Abstract
Gene expression universally relies on protein synthesis, where ribosomes recognize and decode the messenger RNA template by cycling through translation initiation, elongation, and termination phases. All aspects of translation have been studied for decades using the tools of biochemistry and molecular biology available at the time. Here, we focus on the mechanism of translation initiation in eukaryotes, which is remarkably more complex than prokaryotic initiation and is the target of multiple types of regulatory intervention. The "consensus" model, featuring cap-dependent ribosome entry and scanning of mRNA leader sequences, represents the predominantly utilized initiation pathway across eukaryotes, although several variations of the model and alternative initiation mechanisms are also known. Recent advances in structural biology techniques have enabled remarkable molecular-level insights into the functional states of eukaryotic ribosomes, including a range of ribosomal complexes with different combinations of translation initiation factors that are thought to represent bona fide intermediates of the initiation process. Similarly, high-throughput sequencing-based ribosome profiling or "footprinting" approaches have allowed much progress in understanding the elongation phase of translation, and variants of them are beginning to reveal the remaining mysteries of initiation, as well as aspects of translation termination and ribosomal recycling. A current view on the eukaryotic initiation mechanism is presented here with an emphasis on how recent structural and footprinting results underpin axioms of the consensus model. Along the way, we further outline some contested mechanistic issues and major open questions still to be addressed. This article is categorized under: Translation > Translation Mechanisms Translation > Translation Regulation RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Nikolay E Shirokikh
- EMBL-Australia Collaborating Group, Department of Genome Sciences, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Thomas Preiss
- EMBL-Australia Collaborating Group, Department of Genome Sciences, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| |
Collapse
|
150
|
Kramer NJ, Haney MS, Morgens DW, Jovičić A, Couthouis J, Li A, Ousey J, Ma R, Bieri G, Tsui CK, Shi Y, Hertz NT, Tessier-Lavigne M, Ichida JK, Bassik MC, Gitler AD. CRISPR-Cas9 screens in human cells and primary neurons identify modifiers of C9ORF72 dipeptide-repeat-protein toxicity. Nat Genet 2018; 50:603-612. [PMID: 29507424 PMCID: PMC5893388 DOI: 10.1038/s41588-018-0070-7] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 01/24/2018] [Indexed: 12/13/2022]
Abstract
Hexanucleotide repeat expansions in the C9orf72 gene are the most common cause of amyotrophic lateral sclerosis and frontotemporal dementia (c9FTD/ALS). The nucleotide repeat expansions are translated into dipeptide repeat (DPR) proteins, which are aggregation-prone and may contribute to neurodegeneration. We used the CRISPR-Cas9 system to perform genome-wide gene knockout screens for suppressors and enhancers of C9orf72 DPR toxicity in human cells. We validated hits by performing secondary CRISPR-Cas9 screens in primary mouse neurons. We uncovered potent modifiers of DPR toxicity whose gene products function in nucleocytoplasmic transport, the endoplasmic reticulum (ER), proteasome, RNA processing pathways, and in chromatin modification. One modifier, TMX2, modulated the ER-stress signature elicited by C9orf72 DPRs in neurons, and improved survival of human induced motor neurons from C9orf72 ALS patients. Together, this work demonstrates the promise of CRISPR-Cas9 screens to define mechanisms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nicholas J Kramer
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael S Haney
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - David W Morgens
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ana Jovičić
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Molecular Biology, Genentech, South San Francisco, CA, USA
| | - Julien Couthouis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Amy Li
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - James Ousey
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Rosanna Ma
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Gregor Bieri
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - C Kimberly Tsui
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yingxiao Shi
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | | | | | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|