651
|
Dafinca R, Scaber J, Ababneh N, Lalic T, Weir G, Christian H, Vowles J, Douglas AGL, Fletcher-Jones A, Browne C, Nakanishi M, Turner MR, Wade-Martins R, Cowley SA, Talbot K. C9orf72 Hexanucleotide Expansions Are Associated with Altered Endoplasmic Reticulum Calcium Homeostasis and Stress Granule Formation in Induced Pluripotent Stem Cell-Derived Neurons from Patients with Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Stem Cells 2016; 34:2063-78. [PMID: 27097283 PMCID: PMC4979662 DOI: 10.1002/stem.2388] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/26/2016] [Accepted: 03/19/2016] [Indexed: 12/13/2022]
Abstract
An expanded hexanucleotide repeat in a noncoding region of the C9orf72 gene is a major cause of amyotrophic lateral sclerosis (ALS), accounting for up to 40% of familial cases and 7% of sporadic ALS in European populations. We have generated induced pluripotent stem cells (iPSCs) from fibroblasts of patients carrying C9orf72 hexanucleotide expansions, differentiated these to functional motor and cortical neurons, and performed an extensive phenotypic characterization. In C9orf72 iPSC‐derived motor neurons, decreased cell survival is correlated with dysfunction in Ca2+ homeostasis, reduced levels of the antiapoptotic protein Bcl‐2, increased endoplasmic reticulum (ER) stress, and reduced mitochondrial membrane potential. Furthermore, C9orf72 motor neurons, and also cortical neurons, show evidence of abnormal protein aggregation and stress granule formation. This study is an extensive characterization of iPSC‐derived motor neurons as cellular models of ALS carrying C9orf72 hexanucleotide repeats, which describes a novel pathogenic link between C9orf72 mutations, dysregulation of calcium signaling, and altered proteostasis and provides a potential pharmacological target for the treatment of ALS and the related neurodegenerative disease frontotemporal dementia. Stem Cells2016;34:2063–2078
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jane Vowles
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | | | | - Cathy Browne
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Mahito Nakanishi
- Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | | | | | - Sally A Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
652
|
Zhang YJ, Gendron TF, Grima JC, Sasaguri H, Jansen-West K, Xu YF, Katzman RB, Gass J, Murray ME, Shinohara M, Lin WL, Garrett A, Stankowski JN, Daughrity L, Tong J, Perkerson EA, Yue M, Chew J, Castanedes-Casey M, Kurti A, Wang ZS, Liesinger AM, Baker JD, Jiang J, Lagier-Tourenne C, Edbauer D, Cleveland DW, Rademakers R, Boylan KB, Bu G, Link CD, Dickey CA, Rothstein JD, Dickson DW, Fryer JD, Petrucelli L. C9ORF72 poly(GA) aggregates sequester and impair HR23 and nucleocytoplasmic transport proteins. Nat Neurosci 2016; 19:668-677. [PMID: 26998601 PMCID: PMC5138863 DOI: 10.1038/nn.4272] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 02/17/2016] [Indexed: 12/13/2022]
Abstract
Neuronal inclusions of poly(GA), a protein unconventionally translated from G4C2 repeat expansions in C9ORF72, are abundant in patients with frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) caused by this mutation. To investigate poly(GA) toxicity, we generated mice that exhibit poly(GA) pathology, neurodegeneration and behavioral abnormalities reminiscent of FTD and ALS. These phenotypes occurred in the absence of TDP-43 pathology and required poly(GA) aggregation. HR23 proteins involved in proteasomal degradation and proteins involved in nucleocytoplasmic transport were sequestered by poly(GA) in these mice. HR23A and HR23B similarly colocalized to poly(GA) inclusions in C9ORF72 expansion carriers. Sequestration was accompanied by an accumulation of ubiquitinated proteins and decreased xeroderma pigmentosum C (XPC) levels in mice, indicative of HR23A and HR23B dysfunction. Restoring HR23B levels attenuated poly(GA) aggregation and rescued poly(GA)-induced toxicity in neuronal cultures. These data demonstrate that sequestration and impairment of nuclear HR23 and nucleocytoplasmic transport proteins is an outcome of, and a contributor to, poly(GA) pathology.
Collapse
Affiliation(s)
- Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jonathan C Grima
- Department of Neurology, School of Medicine, Johns Hopkins University, Maryland, USA
- Brain Science Institute, School of Medicine, Johns Hopkins University, Maryland, USA
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Maryland, USA
| | - Hiroki Sasaguri
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Ya-Fei Xu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Jennifer Gass
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Wen-Lang Lin
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Aliesha Garrett
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | - Jimei Tong
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Mei Yue
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jeannie Chew
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Zizhao S Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Jeremy D Baker
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, Florida, USA
| | - Jie Jiang
- Ludwig Institute, University of California at San Diego, La Jolla, California, USA
| | | | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Metabolic Biochemistry, Ludwig Maximilians University Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Don W Cleveland
- Ludwig Institute, University of California at San Diego, La Jolla, California, USA
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California, USA
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Kevin B Boylan
- Department of Neurology, Mayo Clinic, Jacksonville, Florida, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Christopher D Link
- Integrative Physiology, Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado, USA
| | - Chad A Dickey
- Department of Molecular Medicine, College of Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, Florida, USA
| | - Jeffrey D Rothstein
- Department of Neurology, School of Medicine, Johns Hopkins University, Maryland, USA
- Brain Science Institute, School of Medicine, Johns Hopkins University, Maryland, USA
- Department of Neuroscience, School of Medicine, Johns Hopkins University, Maryland, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | |
Collapse
|
653
|
C9orf72 BAC Mouse Model with Motor Deficits and Neurodegenerative Features of ALS/FTD. Neuron 2016; 90:521-34. [DOI: 10.1016/j.neuron.2016.04.005] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/01/2016] [Accepted: 03/29/2016] [Indexed: 12/12/2022]
|
654
|
Xiao S, MacNair L, McLean J, McGoldrick P, McKeever P, Soleimani S, Keith J, Zinman L, Rogaeva E, Robertson J. C9orf72 isoforms in Amyotrophic Lateral Sclerosis and Frontotemporal Lobar Degeneration. Brain Res 2016; 1647:43-49. [PMID: 27134035 DOI: 10.1016/j.brainres.2016.04.062] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/23/2016] [Accepted: 04/26/2016] [Indexed: 12/11/2022]
Abstract
A hexanucleotide (G4C2) repeat expansion in the 5' non-coding region C9orf72 is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Three modes of toxicity have been proposed: gain of function through formation of RNA foci and sequestration of RNA binding proteins; expression of dipeptide repeat proteins generated by repeat-associated non-ATG translation; and loss of function due to C9orf72 haploinsufficiency. Much is known about the proposed gain of function mechanisms, but there is little knowledge of the normal function of C9orf72 and the cellular consequences if its activity is perturbed. Here we will review what is known of C9orf72 at the transcript and protein levels and how changes in C9orf72 expression could contribute to disease pathogenesis. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
Affiliation(s)
- Shangxi Xiao
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Canada
| | - Laura MacNair
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Jesse McLean
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Canada
| | - Phillip McGoldrick
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Canada
| | - Paul McKeever
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | - Serena Soleimani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Canada
| | | | | | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Canada
| | - Janice Robertson
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada.
| |
Collapse
|
655
|
Busch JI, Unger TL, Jain N, Tyler Skrinak R, Charan RA, Chen-Plotkin AS. Increased expression of the frontotemporal dementia risk factor TMEM106B causes C9orf72-dependent alterations in lysosomes. Hum Mol Genet 2016; 25:2681-2697. [PMID: 27126638 DOI: 10.1093/hmg/ddw127] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 12/13/2022] Open
Abstract
Frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP) is an important cause of dementia in individuals under age 65. Common variants in the TMEM106B gene were previously discovered by genome-wide association to confer genetic risk for FTLD-TDP (p = 1 × 10-11, OR = 1.6). Furthermore, TMEM106B may act as a genetic modifier affecting age at onset and age at death in the Mendelian subgoup of FTLD-TDP due to expansions of the C9orf72 gene. Evidence suggests that TMEM106B variants increase risk for developing FTLD-TDP by increasing expression of Transmembrane Protein 106B (TMEM106B), a lysosomal protein. To further understand the functional role of TMEM106B in disease pathogenesis, we investigated the cell biological effects of increased TMEM106B expression. Here, we report that increased TMEM106B expression results in the appearance of a vacuolar phenotype in multiple cell types, including neurons. Concomitant with the development of this vacuolar phenotype, cells over-expressing TMEM106B exhibit impaired lysosomal acidification and degradative function, as well as increased cytotoxicity. We further identify a potential lysosomal sorting motif for TMEM106B and demonstrate that abrogation of sorting to lysosomes rescues TMEM106B-induced defects. Finally, we show that TMEM106B-induced defects are dependent on the presence of C9orf72, as knockdown of C9orf72 also rescues these defects. In sum, our results suggest that TMEM106B exerts its effects on FTLD-TDP disease risk through alterations in lysosomal pathways. Furthermore, TMEM106B and C9orf72 may interact in FTLD-TDP pathophysiology.
Collapse
Affiliation(s)
- Johanna I Busch
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Travis L Unger
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nimansha Jain
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - R Tyler Skrinak
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rakshita A Charan
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
656
|
Sellier C, Campanari ML, Julie Corbier C, Gaucherot A, Kolb-Cheynel I, Oulad-Abdelghani M, Ruffenach F, Page A, Ciura S, Kabashi E, Charlet-Berguerand N. Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J 2016; 35:1276-97. [PMID: 27103069 DOI: 10.15252/embj.201593350] [Citation(s) in RCA: 318] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 03/15/2016] [Indexed: 12/12/2022] Open
Abstract
An intronic expansion of GGGGCC repeats within the C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD). Ataxin-2 with intermediate length of polyglutamine expansions (Ataxin-2 Q30x) is a genetic modifier of the disease. Here, we found that C9ORF72 forms a complex with the WDR41 and SMCR8 proteins to act as a GDP/GTP exchange factor for RAB8a and RAB39b and to thereby control autophagic flux. Depletion of C9orf72 in neurons partly impairs autophagy and leads to accumulation of aggregates of TDP-43 and P62 proteins, which are histopathological hallmarks of ALS-FTD SMCR8 is phosphorylated by TBK1 and depletion of TBK1 can be rescued by phosphomimetic mutants of SMCR8 or by constitutively active RAB39b, suggesting that TBK1, SMCR8, C9ORF72, and RAB39b belong to a common pathway regulating autophagy. While depletion of C9ORF72 only has a partial deleterious effect on neuron survival, it synergizes with Ataxin-2 Q30x toxicity to induce motor neuron dysfunction and neuronal cell death. These results indicate that partial loss of function of C9ORF72 is not deleterious by itself but synergizes with Ataxin-2 toxicity, suggesting a double-hit pathological mechanism in ALS-FTD.
Collapse
Affiliation(s)
- Chantal Sellier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Maria-Letizia Campanari
- Sorbonne Université, Université Pierre et Marie Curie (UPMC), Université de Paris 06, Unité Mixte 75, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1127, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225 Institut du Cerveau et de la Moelle Épinière (ICM), 75013, Paris, France
| | - Camille Julie Corbier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Angeline Gaucherot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Isabelle Kolb-Cheynel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Mustapha Oulad-Abdelghani
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Frank Ruffenach
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Adeline Page
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Sorana Ciura
- Sorbonne Université, Université Pierre et Marie Curie (UPMC), Université de Paris 06, Unité Mixte 75, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1127, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225 Institut du Cerveau et de la Moelle Épinière (ICM), 75013, Paris, France
| | - Edor Kabashi
- Sorbonne Université, Université Pierre et Marie Curie (UPMC), Université de Paris 06, Unité Mixte 75, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1127, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225 Institut du Cerveau et de la Moelle Épinière (ICM), 75013, Paris, France
| | - Nicolas Charlet-Berguerand
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| |
Collapse
|
657
|
Rizzu P, Blauwendraat C, Heetveld S, Lynes EM, Castillo-Lizardo M, Dhingra A, Pyz E, Hobert M, Synofzik M, Simón-Sánchez J, Francescatto M, Heutink P. C9orf72 is differentially expressed in the central nervous system and myeloid cells and consistently reduced in C9orf72, MAPT and GRN mutation carriers. Acta Neuropathol Commun 2016; 4:37. [PMID: 27079381 PMCID: PMC4832459 DOI: 10.1186/s40478-016-0306-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 03/25/2016] [Indexed: 12/12/2022] Open
Abstract
A non-coding hexanucleotide repeat expansion (HRE) in C9orf72 is a common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) acting through a loss of function mechanism due to haploinsufficiency of C9orf72 or a gain of function mediated by aggregates of bidirectionally transcribed HRE-RNAs translated into di-peptide repeat (DPR) proteins. To fully understand regulation of C9orf72 expression we surveyed the C9orf72 locus using Cap Analysis of Gene Expression sequence data (CAGEseq). We observed C9orf72 was generally lowly expressed with the exception of a subset of myeloid cells, particularly CD14+ monocytes that showed up to seven fold higher expression as compared to central nervous system (CNS) and other tissues. The expression profile at the C9orf72 locus showed a complex architecture with differential expression of the transcription start sites (TSSs) for the annotated C9orf72 transcripts between myeloid and CNS tissues suggesting cell and/or tissue specific functions. We further detected novel TSSs in both the sense and antisense strand at the C9orf72 locus and confirmed their existence in brain tissues and CD14+ monocytes. Interestingly, our experiments showed a consistent decrease of C9orf72 coding transcripts not only in brain tissue and monocytes from C9orf72-HRE patients, but also in brains from MAPT and GRN mutation carriers together with an increase in antisense transcripts suggesting these could play a role in regulation of C9orf72. We found that the non-HRE related expression changes cannot be explained by promoter methylation but by the presence of the C9orf72-HRE risk haplotype and unknown functional interactions between C9orf72, MAPT and GRN.
Collapse
|
658
|
Protein folding alterations in amyotrophic lateral sclerosis. Brain Res 2016; 1648:633-649. [PMID: 27064076 DOI: 10.1016/j.brainres.2016.04.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/04/2016] [Accepted: 04/05/2016] [Indexed: 02/07/2023]
Abstract
Protein misfolding leads to the formation of aggregated proteins and protein inclusions, which are associated with synaptic loss and neuronal death in neurodegenerative diseases. Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that targets motor neurons in the brain, brainstem and spinal cord. Several proteins misfold and are associated either genetically or pathologically in ALS, including superoxide dismutase 1 (SOD1), Tar DNA binding protein-43 (TDP-43), Ubiquilin-2, p62, VCP, and dipeptide repeat proteins produced by unconventional repeat associated non-ATG translation of the repeat expansion in C9ORF72. Chaperone proteins, including heat shock proteins (Hsp׳s) and the protein disulphide isomerase (PDI) family, assist in protein folding and therefore can prevent protein misfolding, and have been implicated as being protective in ALS. In this review we provide an overview of the current literature regarding the molecular mechanisms of protein misfolding and aggregation in ALS, and the role of chaperones as potential targets for therapeutic intervention. This article is part of a Special Issue entitled SI:ER stress.
Collapse
|
659
|
Green KM, Linsalata AE, Todd PK. RAN translation-What makes it run? Brain Res 2016; 1647:30-42. [PMID: 27060770 DOI: 10.1016/j.brainres.2016.04.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/24/2016] [Accepted: 04/01/2016] [Indexed: 12/14/2022]
Abstract
Nucleotide-repeat expansions underlie a heterogeneous group of neurodegenerative and neuromuscular disorders for which there are currently no effective therapies. Recently, it was discovered that such repetitive RNA motifs can support translation initiation in the absence of an AUG start codon across a wide variety of sequence contexts, and that the products of these atypical translation initiation events contribute to neuronal toxicity. This review examines what we currently know and do not know about repeat associated non-AUG (RAN) translation in the context of established canonical and non-canonical mechanisms of translation initiation. We highlight recent findings related to RAN translation in three repeat expansion disorders: CGG repeats in fragile X-associated tremor ataxia syndrome (FXTAS), GGGGCC repeats in C9orf72 associated amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) and CAG repeats in Huntington disease. These studies suggest that mechanistic differences may exist for RAN translation dependent on repeat type, repeat reading frame, and the surrounding sequence context, but that for at least some repeats, RAN translation retains a dependence on some of the canonical translational initiation machinery. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
Affiliation(s)
- Katelyn M Green
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Alexander E Linsalata
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Peter K Todd
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Veterans Affairs Medical Center, Ann Arbor, MI, United States.
| |
Collapse
|
660
|
Gitler AD, Tsuiji H. There has been an awakening: Emerging mechanisms of C9orf72 mutations in FTD/ALS. Brain Res 2016; 1647:19-29. [PMID: 27059391 DOI: 10.1016/j.brainres.2016.04.004] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/30/2016] [Accepted: 04/03/2016] [Indexed: 12/13/2022]
Abstract
The discovery of C9orf72 mutations as the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) has awakened a surge of interest in deciphering how mutations in this mysterious gene cause disease and what can be done to stop it. C9orf72 harbors a hexanucleotide repeat, GGGGCC, in a non-coding region of the gene and a massive expansion of this repeat causes ALS, FTD, or both (FTD/ALS). Many questions lie ahead. What does this gene normally do? What is the consequence of an enormous GGGGCC repeat expansion on that gene's function? Could that hexanucleotide repeat expansion have additional pathological actions unrelated to C9orf72 function? There has been tremendous progress on all fronts in the quest to define how C9orf72 mutations cause disease. Many new experimental models have been constructed and unleashed in powerful genetic screens. Studies in mouse and human patient samples, including iPS-derived neurons, have provided unprecedented insights into pathogenic mechanisms. Three major hypotheses have emerged and are still being hotly debated in the field. These include (1) loss of function owing to decrease in the abundance of C9orf72 protein and its ability to carryout its still unknown cellular role; (2) RNA toxicity from bidirectionally transcribed sense (GGGGCC) and antisense (GGCCCC) transcripts that accumulate in RNA foci and might sequester critical RNA-binding proteins; (3) proteotoxicity from dipeptide repeat proteins produced by an unconventional form of translation from the expanded nucleotide repeats. Here we review the evidence in favor and against each of these three hypotheses. We also suggest additional experiments and considerations that we propose will help clarify which mechanism(s) are most important for driving disease and therefore most critical for considering during the development of therapeutic interventions. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
Affiliation(s)
- Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, 300 Pasteur Drive, M322 Alway Building, Stanford, CA 94305, USA.
| | - Hitomi Tsuiji
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori, Mizuhoku, Nagoya, Aichi 467-8603, Japan.
| |
Collapse
|
661
|
C9ORF72 Regulates Stress Granule Formation and Its Deficiency Impairs Stress Granule Assembly, Hypersensitizing Cells to Stress. Mol Neurobiol 2016; 54:3062-3077. [DOI: 10.1007/s12035-016-9850-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 03/09/2016] [Indexed: 12/14/2022]
|
662
|
Kearse MG, Green KM, Krans A, Rodriguez CM, Linsalata AE, Goldstrohm AC, Todd PK. CGG Repeat-Associated Non-AUG Translation Utilizes a Cap-Dependent Scanning Mechanism of Initiation to Produce Toxic Proteins. Mol Cell 2016; 62:314-322. [PMID: 27041225 DOI: 10.1016/j.molcel.2016.02.034] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/25/2016] [Accepted: 02/26/2016] [Indexed: 02/03/2023]
Abstract
Repeat-associated non-AUG (RAN) translation produces toxic polypeptides from nucleotide repeat expansions in the absence of an AUG start codon and contributes to neurodegenerative disorders such as ALS and fragile X-associated tremor/ataxia syndrome. How RAN translation occurs is unknown. Here we define the critical sequence and initiation factors that mediate CGG repeat RAN translation in the 5' leader of fragile X mRNA, FMR1. Our results reveal that CGG RAN translation is 30%-40% as efficient as AUG-initiated translation, is m(7)G cap and eIF4E dependent, requires the eIF4A helicase, and is strongly influenced by repeat length. However, it displays a dichotomous requirement for initiation site selection between reading frames, with initiation in the +1 frame, but not the +2 frame, occurring at near-cognate start codons upstream of the repeat. These data support a model in which RAN translation at CGG repeats uses cap-dependent ribosomal scanning, yet bypasses normal requirements for start codon selection.
Collapse
Affiliation(s)
- Michael G Kearse
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Katelyn M Green
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Amy Krans
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Veterans Affairs Medical Center, Ann Arbor, MI 48105, USA
| | - Caitlin M Rodriguez
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alexander E Linsalata
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Aaron C Goldstrohm
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Peter K Todd
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Veterans Affairs Medical Center, Ann Arbor, MI 48105, USA; Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
663
|
Ruegsegger C, Saxena S. Proteostasis impairment in ALS. Brain Res 2016; 1648:571-579. [PMID: 27033833 DOI: 10.1016/j.brainres.2016.03.032] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 03/20/2016] [Accepted: 03/21/2016] [Indexed: 12/13/2022]
Abstract
In physiological conditions the maintenance of the cellular proteome is a prerequisite for optimal cell functioning and cell survival. Additionally, cells need to constantly sense and adapt to their changing environment and associated stressors. Cells achieve this via a set of molecular chaperones, protein clearance pathways as well as stress-associated signaling networks which work together to prevent protein misfolding, its aggregation and accumulation in subcellular compartments. These processes together form the proteostasis network which helps in maintaining cellular proteostasis. Imbalance or impairment in this processes is directly linked to ageing associated disorders such as diabetes, cancer, stroke, metabolic disorders, pulmonary fibrosis, inflammation and neurodegenerative diseases. In this review, we provide insights into the proteostasis process and how its failure governs neurodegenerative disorders with a special focus on Amyotrophic lateral sclerosis (ALS). This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- Céline Ruegsegger
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, CH-3012 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Smita Saxena
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, CH-3012 Bern, Switzerland.
| |
Collapse
|
664
|
O'Rourke JG, Bogdanik L, Muhammad AKMG, Gendron TF, Kim KJ, Austin A, Cady J, Liu EY, Zarrow J, Grant S, Ho R, Bell S, Carmona S, Simpkinson M, Lall D, Wu K, Daughrity L, Dickson DW, Harms MB, Petrucelli L, Lee EB, Lutz CM, Baloh RH. C9orf72 BAC Transgenic Mice Display Typical Pathologic Features of ALS/FTD. Neuron 2016; 88:892-901. [PMID: 26637796 DOI: 10.1016/j.neuron.2015.10.027] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 06/16/2015] [Accepted: 09/09/2015] [Indexed: 12/13/2022]
Abstract
Noncoding expansions of a hexanucleotide repeat (GGGGCC) in the C9orf72 gene are the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia. Here we report transgenic mice carrying a bacterial artificial chromosome (BAC) containing the full human C9orf72 gene with either a normal allele (15 repeats) or disease-associated expansion (∼100-1,000 repeats; C9-BACexp). C9-BACexp mice displayed pathologic features seen in C9orf72 expansion patients, including widespread RNA foci and repeat-associated non-ATG (RAN) translated dipeptides, which were suppressed by antisense oligonucleotides targeting human C9orf72. Nucleolin distribution was altered, supporting that either C9orf72 transcripts or RAN dipeptides promote nucleolar dysfunction. Despite early and widespread production of RNA foci and RAN dipeptides in C9-BACexp mice, behavioral abnormalities and neurodegeneration were not observed even at advanced ages, supporting the hypothesis that RNA foci and RAN dipeptides occur presymptomatically and are not sufficient to drive neurodegeneration in mice at levels seen in patients.
Collapse
Affiliation(s)
- Jacqueline G O'Rourke
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Laurent Bogdanik
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - A K M G Muhammad
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Kevin J Kim
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Andrew Austin
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Janet Cady
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Elaine Y Liu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonah Zarrow
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Sharday Grant
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Ritchie Ho
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Shaughn Bell
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Sharon Carmona
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Megan Simpkinson
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Deepti Lall
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Kathryn Wu
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Lillian Daughrity
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Matthew B Harms
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cathleen M Lutz
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Robert H Baloh
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; Department of Neurology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA.
| |
Collapse
|
665
|
Atanasio A, Decman V, White D, Ramos M, Ikiz B, Lee HC, Siao CJ, Brydges S, LaRosa E, Bai Y, Fury W, Burfeind P, Zamfirova R, Warshaw G, Orengo J, Oyejide A, Fralish M, Auerbach W, Poueymirou W, Freudenberg J, Gong G, Zambrowicz B, Valenzuela D, Yancopoulos G, Murphy A, Thurston G, Lai KMV. C9orf72 ablation causes immune dysregulation characterized by leukocyte expansion, autoantibody production, and glomerulonephropathy in mice. Sci Rep 2016; 6:23204. [PMID: 26979938 PMCID: PMC4793236 DOI: 10.1038/srep23204] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/02/2016] [Indexed: 12/12/2022] Open
Abstract
The expansion of a hexanucleotide (GGGGCC) repeat in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Both the function of C9ORF72 and the mechanism by which the repeat expansion drives neuropathology are unknown. To examine whether C9ORF72 haploinsufficiency induces neurological disease, we created a C9orf72-deficient mouse line. Null mice developed a robust immune phenotype characterized by myeloid expansion, T cell activation, and increased plasma cells. Mice also presented with elevated autoantibodies and evidence of immune-mediated glomerulonephropathy. Collectively, our data suggest that C9orf72 regulates immune homeostasis and an autoimmune response reminiscent of systemic lupus erythematosus (SLE) occurs in its absence. We further imply that haploinsufficiency is unlikely to be the causative factor in C9ALS/FTD pathology.
Collapse
Affiliation(s)
| | - Vilma Decman
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY USA
| | - Derek White
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY USA
| | - Meg Ramos
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY USA
| | - Burcin Ikiz
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY USA
| | | | | | | | | | - Yu Bai
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY USA
| | - Wen Fury
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY USA
| | | | | | | | - Jamie Orengo
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY USA
| | | | | | | | | | | | - Guochun Gong
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY USA
| | | | | | | | | | | | | |
Collapse
|
666
|
Xia G, Gao Y, Jin S, Subramony SH, Terada N, Ranum LPW, Swanson MS, Ashizawa T. Genome modification leads to phenotype reversal in human myotonic dystrophy type 1 induced pluripotent stem cell-derived neural stem cells. Stem Cells 2016; 33:1829-38. [PMID: 25702800 DOI: 10.1002/stem.1970] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 01/17/2015] [Indexed: 12/15/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is caused by expanded CTG repeats in the 3'-untranslated region (3' UTR) of the DMPK gene. Correcting the mutation in DM1 stem cells would be an important step toward autologous stem cell therapy. The objective of this study is to demonstrate in vitro genome editing to prevent production of toxic mutant transcripts and reverse phenotypes in DM1 stem cells. Genome editing was performed in DM1 neural stem cells (NSCs) derived from human DM1 induced pluripotent stem (iPS) cells. An editing cassette containing SV40/bGH polyA signals was integrated upstream of the CTG repeats by TALEN-mediated homologous recombination (HR). The expression of mutant CUG repeats transcript was monitored by nuclear RNA foci, the molecular hallmarks of DM1, using RNA fluorescence in situ hybridization. Alternative splicing of microtubule-associated protein tau (MAPT) and muscleblind-like (MBNL) proteins were analyzed to further monitor the phenotype reversal after genome modification. The cassette was successfully inserted into DMPK intron 9 and this genomic modification led to complete disappearance of nuclear RNA foci. MAPT and MBNL 1, 2 aberrant splicing in DM1 NSCs were reversed to normal pattern in genome-modified NSCs. Genome modification by integration of exogenous polyA signals upstream of the DMPK CTG repeat expansion prevents the production of toxic RNA and leads to phenotype reversal in human DM1 iPS-cells derived stem cells. Our data provide proof-of-principle evidence that genome modification may be used to generate genetically modified progenitor cells as a first step toward autologous cell transfer therapy for DM1.
Collapse
Affiliation(s)
- Guangbin Xia
- Department of Neurology, University of Florida, College of Medicine, Gainesville, Florida, USA.,Center for Cellular Reprogramming, University of Florida, College of Medicine, Gainesville, Florida, USA.,Center for NeuroGenetics, University of Florida, College of Medicine, Gainesville, Florida, USA.,The Evelyn L & William F. McKnight Brain Institute, University of Florida, Gainesville, Florida, USA.,Department of Neuroscience, University of Florida, Gainesville, Florida, USA
| | - Yuanzheng Gao
- Department of Neurology, University of Florida, College of Medicine, Gainesville, Florida, USA.,The Evelyn L & William F. McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Shouguang Jin
- Department of Molecular Genetics and Microbiology, College of Medicine, Gainesville, Florida, USA
| | - S H Subramony
- Department of Neurology, University of Florida, College of Medicine, Gainesville, Florida, USA.,Center for NeuroGenetics, University of Florida, College of Medicine, Gainesville, Florida, USA.,The Evelyn L & William F. McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Naohiro Terada
- Center for Cellular Reprogramming, University of Florida, College of Medicine, Gainesville, Florida, USA.,Department of Pathology, Immunology & Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Laura P W Ranum
- Department of Neurology, University of Florida, College of Medicine, Gainesville, Florida, USA.,Center for NeuroGenetics, University of Florida, College of Medicine, Gainesville, Florida, USA.,Department of Molecular Genetics and Microbiology, College of Medicine, Gainesville, Florida, USA.,Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Maurice S Swanson
- Center for NeuroGenetics, University of Florida, College of Medicine, Gainesville, Florida, USA.,Department of Molecular Genetics and Microbiology, College of Medicine, Gainesville, Florida, USA.,Genetics Institute, University of Florida, Gainesville, Florida, USA
| | - Tetsuo Ashizawa
- Department of Neurology, University of Florida, College of Medicine, Gainesville, Florida, USA.,Center for Cellular Reprogramming, University of Florida, College of Medicine, Gainesville, Florida, USA.,Center for NeuroGenetics, University of Florida, College of Medicine, Gainesville, Florida, USA.,The Evelyn L & William F. McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
667
|
Geevasinga N, Menon P, Nicholson GA, Ng K, Howells J, Kril JJ, Yiannikas C, Kiernan MC, Vucic S. Cortical Function in Asymptomatic Carriers and Patients With C9orf72 Amyotrophic Lateral Sclerosis. JAMA Neurol 2016; 72:1268-74. [PMID: 26348842 DOI: 10.1001/jamaneurol.2015.1872] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IMPORTANCE The identification of the chromosome 9 open reading frame 72 (c9orf72) gene hexanucleotide repeat expansion represents a major advance in the understanding of amyotrophic lateral sclerosis (ALS) pathogenesis. The pathophysiological mechanism by which the c9orf72 gene expansion leads to neurodegeneration is not yet elucidated. Cortical hyperexcitability is potentially an important pathophysiological process in sporadic ALS and familial ALS (FALS). OBJECTIVE To investigate whether cortical hyperexcitability forms the pathophysiological basis of c9orf72 FALS using the threshold-tracking transcranial magnetic stimulation technique. DESIGN, SETTING, AND PARTICIPANTS Prospective case-control single-center study that took place at hospitals and outpatient clinics from January 1, 2013, to January 1, 2015. Clinical and functional assessments along with transcranial magnetic stimulation studies were taken on 15 patients with c9orf72 FALS and 11 asymptomatic expansion carriers of c9orf72 who were longitudinally followed up for 3 years. Results were compared with 73 patients with sporadic ALS and 74 healthy control participants. MAIN OUTCOMES AND MEASURES Cortical excitability variables, including short-interval intracortical inhibition, were measured in patients with c9orf72 FALS and results were compared with asymptomatic c9orf72 carriers, patients with sporadic ALS, and healthy control participants. RESULTS Mean (SD) short-interval intracortical inhibition was significantly reduced in patients with c9orf72 FALS (1.2% [1.8%]) and sporadic ALS (1.6% [1.2%]) compared with asymptomatic c9orf72 expansion carriers (10.2% [1.8%]; F = 16.1; P < .001) and healthy control participants (11.8% [1.0%]; F = 16.1; P < .001). The reduction of short-interval intracortical inhibition was accompanied by an increase in intracortical facilitation (P < .01) and motor-evoked potential amplitude (P < .05) as well as a reduction in the resting motor threshold (P < .05) and cortical silent period duration (P < .001). CONCLUSIONS AND RELEVANCE This study establishes cortical hyperexcitability as an intrinsic feature of symptomatic c9orf72 expansion-related ALS but not asymptomatic expansion carriers.
Collapse
Affiliation(s)
- Nimeshan Geevasinga
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Parvathi Menon
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Garth A Nicholson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, New South Wales, Australia
| | - Karl Ng
- Department of Neurology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - James Howells
- Brain and Mind Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Jillian J Kril
- Disciplines of Medicine and Pathology, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Con Yiannikas
- Department of Neurology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Matthew C Kiernan
- Brain and Mind Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Steve Vucic
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
668
|
Bañez-Coronel M, Ayhan F, Tarabochia AD, Zu T, Perez BA, Tusi SK, Pletnikova O, Borchelt DR, Ross CA, Margolis RL, Yachnis AT, Troncoso JC, Ranum LPW. RAN Translation in Huntington Disease. Neuron 2016; 88:667-77. [PMID: 26590344 DOI: 10.1016/j.neuron.2015.10.038] [Citation(s) in RCA: 266] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 09/05/2015] [Accepted: 10/15/2015] [Indexed: 11/30/2022]
Abstract
Huntington disease (HD) is caused by a CAG ⋅ CTG expansion in the huntingtin (HTT) gene. While most research has focused on the HTT polyGln-expansion protein, we demonstrate that four additional, novel, homopolymeric expansion proteins (polyAla, polySer, polyLeu, and polyCys) accumulate in HD human brains. These sense and antisense repeat-associated non-ATG (RAN) translation proteins accumulate most abundantly in brain regions with neuronal loss, microglial activation and apoptosis, including caudate/putamen, white matter, and, in juvenile-onset cases, also the cerebellum. RAN protein accumulation and aggregation are length dependent, and individual RAN proteins are toxic to neural cells independent of RNA effects. These data suggest RAN proteins contribute to HD and that therapeutic strategies targeting both sense and antisense genes may be required for efficacy in HD patients. This is the first demonstration that RAN proteins are expressed across an expansion located in an open reading frame and suggests RAN translation may also contribute to other polyglutamine diseases.
Collapse
Affiliation(s)
- Monica Bañez-Coronel
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, USA; Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Fatma Ayhan
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, USA; Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Alex D Tarabochia
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, USA; Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Tao Zu
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, USA; Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Barbara A Perez
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, USA; Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Solaleh Khoramian Tusi
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, USA; Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA; Genetics Institute, University of Florida, Gainesville, FL 32610, USA
| | - Olga Pletnikova
- Department of Pathology, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David R Borchelt
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| | - Christopher A Ross
- Division of Neurobiology, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Program in Cellular and Molecular Medicine, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Baltimore Huntington's Disease Center, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Russell L Margolis
- Division of Neurobiology, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Baltimore Huntington's Disease Center, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anthony T Yachnis
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Juan C Troncoso
- Department of Pathology, The John Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, The John Hopkins University School of Medicine, Baltimore, MD 21205, 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, University of Florida, Gainesville, FL 32610, USA; Genetics Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
669
|
Niblock M, Smith BN, Lee YB, Sardone V, Topp S, Troakes C, Al-Sarraj S, Leblond CS, Dion PA, Rouleau GA, Shaw CE, Gallo JM. Retention of hexanucleotide repeat-containing intron in C9orf72 mRNA: implications for the pathogenesis of ALS/FTD. Acta Neuropathol Commun 2016; 4:18. [PMID: 26916632 PMCID: PMC4766718 DOI: 10.1186/s40478-016-0289-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 02/15/2016] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION The most common forms of amyotrophic lateral sclerosis and frontotemporal dementia are caused by a large GGGGCC repeat expansion in the first intron of the C9orf72 gene. The repeat-containing intron should be degraded after being spliced out, however GGGGCC repeat-containing RNA species either accumulate in nuclear foci or are exported to the cytoplasm where they are translated into potentially toxic dipeptide repeat proteins by repeat-associated non-AUG-initiated (RAN) translation. RESULTS In order to determine the mechanisms of repeat-containing intron misprocessing, we have analyzed C9orf72 transcripts in lymphoblasts from C9orf72 expansion carriers (n = 15) and control individuals (n = 15). We have identified polyadenylated C9orf72 RNA species retaining the repeat-containing intron and in which downstream exons are spliced correctly resulting in a C9orf72 mRNA with an enlarged 5'-UTR containing the GGGGCC repeats. Intron-retaining transcripts are produced from both wild-type and mutant alleles. Intron-retaining C9orf72 transcripts were also detected in brain with a 2.7 fold increase measured in the frontal cortex from heterozygous expansion carriers (n = 11) compared to controls (n = 10). The level of intron-retaining transcripts was increased 5.9 fold in a case homozygous for the expansion. We also show that a large proportion of intron 1-retaining C9orf72 transcripts accumulate in the nucleus. CONCLUSIONS Retention of the repeat-containing intron in mature C9orf72 mRNA can potentially explain nuclear foci formation as well as nuclear export of GGGGCC repeat RNA and suggests that the misprocessing of C9orf72 transcripts initiates the pathogenic process caused by C9orf72 hexanucleotide repeat expansions as well as provides the basis for novel therapeutic strategies.
Collapse
Affiliation(s)
- Michael Niblock
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Bradley N Smith
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Youn-Bok Lee
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Valentina Sardone
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Simon Topp
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
- London Neurodegenerative Disease Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Safa Al-Sarraj
- London Neurodegenerative Disease Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Claire S Leblond
- Montreal Neurological Institute and Hospital, McGill University, Ludmer Building, 1033 Pine Avenue West, Montreal, QC, H3A 1A1, Canada
| | - Patrick A Dion
- Montreal Neurological Institute and Hospital, McGill University, Ludmer Building, 1033 Pine Avenue West, Montreal, QC, H3A 1A1, Canada
| | - Guy A Rouleau
- Montreal Neurological Institute and Hospital, McGill University, Ludmer Building, 1033 Pine Avenue West, Montreal, QC, H3A 1A1, Canada
| | - Christopher E Shaw
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Jean-Marc Gallo
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
670
|
Ishiguro A, Kimura N, Watanabe Y, Watanabe S, Ishihama A. TDP-43 binds and transports G-quadruplex-containing mRNAs into neurites for local translation. Genes Cells 2016; 21:466-81. [PMID: 26915990 DOI: 10.1111/gtc.12352] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 02/01/2016] [Indexed: 12/14/2022]
Abstract
Growth and differentiation of the neurites depends on long-distance transport of a specific set of mRNAs to restricted area and their local translation. Here, we found that a TAR DNA-binding protein of 43 kDa in size (TDP-43) plays an essential role in intracellular transport of mRNA. For identification of target RNAs recognized by TDP-43, we purified TDP-43 in soluble dimer form and subjected to in vitro systematic evolution of ligands by exponential enrichment (SELEX) screening. All the TDP-43-bound RNAs were found to contain G-quadruplex (G4). Using a double-fluorescent probe system, G4-containing RNAs were found to be transported, together with TDP-43, into the distal neurites. Two lines of evidence indicated that loss of function of TDP-43 results in the neurodegenerative disorder: (i) amyotrophic lateral sclerosis (ALS)-linked mutant TDP-43M337V lacks the activity of binding and transport of G4-containing mRNAs; and (ii) RNA containing G4-forming GGGGCC repeat expansion from the ALS-linked C9orf72 gene absorbs TDP-43, thereby reducing the intracellular pool of functional TDP-43. Taken together, we propose that TDP-43 within neurons plays an essential role of mRNA transport into distal neurites for local translation, and thus, dysfunctions of TDP-43 cause neural diseases such as ALS and frontotemporal lobar degeneration.
Collapse
Affiliation(s)
- Akira Ishiguro
- Research Center for Micro-Nano Technology, University of Hosei, Midori-cho 3-11-15, Koganei, Tokyo, 184-0003, Japan
| | - Nobuyuki Kimura
- Section of Cell Biology and Pathology, Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Morioka 7-430, Obu, Aichi, 474-8511, Japan
| | - Yuto Watanabe
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Yayoi 2-11-16, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Sumiko Watanabe
- Department of Molecular and Developmental Biology, Institute of Medical Science, The University of Tokyo, Shirokane-dai 4-6-1, Minatoku-ku, Tokyo, 108-8639, Japan
| | - Akira Ishihama
- Research Center for Micro-Nano Technology, University of Hosei, Midori-cho 3-11-15, Koganei, Tokyo, 184-0003, Japan
| |
Collapse
|
671
|
Boeynaems S, Bogaert E, Michiels E, Gijselinck I, Sieben A, Jovičić A, De Baets G, Scheveneels W, Steyaert J, Cuijt I, Verstrepen KJ, Callaerts P, Rousseau F, Schymkowitz J, Cruts M, Van Broeckhoven C, Van Damme P, Gitler AD, Robberecht W, Van Den Bosch L. Drosophila screen connects nuclear transport genes to DPR pathology in c9ALS/FTD. Sci Rep 2016; 6:20877. [PMID: 26869068 PMCID: PMC4751451 DOI: 10.1038/srep20877] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 01/11/2016] [Indexed: 12/12/2022] Open
Abstract
Hexanucleotide repeat expansions in C9orf72 are the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD) (c9ALS/FTD). Unconventional translation of these repeats produces dipeptide repeat proteins (DPRs) that may cause neurodegeneration. We performed a modifier screen in Drosophila and discovered a critical role for importins and exportins, Ran-GTP cycle regulators, nuclear pore components, and arginine methylases in mediating DPR toxicity. These findings provide evidence for an important role for nucleocytoplasmic transport in the pathogenic mechanism of c9ALS/FTD.
Collapse
Affiliation(s)
- Steven Boeynaems
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium.,VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| | - Elke Bogaert
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium.,VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| | - Emiel Michiels
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium.,VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| | - Ilse Gijselinck
- Department of Molecular Genetics, VIB, B-2610 Antwerp, Belgium.,Institute Born-Bunge, University of Antwerp, B-2610 Antwerp, Belgium
| | - Anne Sieben
- Department of Molecular Genetics, VIB, B-2610 Antwerp, Belgium.,Institute Born-Bunge, University of Antwerp, B-2610 Antwerp, Belgium.,Department of Neurology, University Hospital Ghent and University of Ghent, B-9000 Ghent, Belgium
| | - Ana Jovičić
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Greet De Baets
- Switch Laboratory, VIB, B-3000 Leuven, Belgium.,Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Wendy Scheveneels
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium.,VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| | - Jolien Steyaert
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium.,VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| | - Ivy Cuijt
- Department of Molecular Genetics, VIB, B-2610 Antwerp, Belgium.,Institute Born-Bunge, University of Antwerp, B-2610 Antwerp, Belgium
| | - Kevin J Verstrepen
- VIB Laboratory of Systems Biology, Gaston Geenslaan 1, B-3001 Leuven, Belgium.,KU Leuven - University of Leuven, Department of Microbial and Molecular Systems, Laboratory for Genetics and Genomics, Gaston Geenslaan 1, B-3001 Leuven, Belgium
| | - Patrick Callaerts
- KU Leuven - University of Leuven, Department of Human Genetics, Laboratory of Behavioral and Developmental Genetics, B-3000 Leuven, Belgium.,VIB Center for the Biology of Disease, B-3000 Leuven, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB, B-3000 Leuven, Belgium.,Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB, B-3000 Leuven, Belgium.,Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Marc Cruts
- Department of Molecular Genetics, VIB, B-2610 Antwerp, Belgium.,Institute Born-Bunge, University of Antwerp, B-2610 Antwerp, Belgium
| | - Christine Van Broeckhoven
- Department of Molecular Genetics, VIB, B-2610 Antwerp, Belgium.,Institute Born-Bunge, University of Antwerp, B-2610 Antwerp, Belgium
| | - Philip Van Damme
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium.,VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium.,University Hospitals Leuven, Department of Neurology, B-3000 Leuven, Belgium
| | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Wim Robberecht
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium.,VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium.,University Hospitals Leuven, Department of Neurology, B-3000 Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Research Institute for Neuroscience and Disease (LIND), B-3000 Leuven, Belgium.,VIB, Vesalius Research Center, Laboratory of Neurobiology, B-3000 Leuven, Belgium
| |
Collapse
|
672
|
An amyloid-like cascade hypothesis for C9orf72 ALS/FTD. Curr Opin Neurobiol 2016; 36:99-106. [DOI: 10.1016/j.conb.2015.10.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/08/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
|
673
|
Eitan C, Hornstein E. Vulnerability of microRNA biogenesis in FTD-ALS. Brain Res 2016; 1647:105-111. [PMID: 26778173 DOI: 10.1016/j.brainres.2015.12.063] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 12/29/2015] [Accepted: 12/31/2015] [Indexed: 12/12/2022]
Abstract
The genetics of the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) turn our attention to RNA metabolism, primarily because many of the identified diseases-associated genes encode for RNA-binding proteins. microRNAs (miRNAs) are endogenous noncoding RNAs that play critical roles in maintaining brain integrity. The current review sheds light on miRNA dysregulation in neurodegenerative diseases, focusing on FTD-ALS. We propose that miRNAs are susceptible to fail when protein factors that are critical for miRNA biogenesis malfunction. Accordingly, potential insufficiencies of the 'microprocessor' complex, the nucleo-cytoplasmic export of miRNA precursors or their processing by Dicer were recently reported. Furthermore, specific miRNAs are involved in the regulation of pathways that are essential for neuronal survival or function. Any change in the expression of these specific miRNAs or in their ability to recognize their target sequences will have negative consequences. Taken together, recent reports strengthens the hypothesis that dysregulation of miRNAs might play an important role in the pathogenesis of neurodegenerative diseases, and highlights the miRNA biogenesis machinery as an interesting target for therapeutic interventions for ALS as well as FTD. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
Affiliation(s)
- Chen Eitan
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Hornstein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
674
|
Tsubota A, Ichijo H, Homma K. Mislocalization, aggregation formation and defect in proteolysis in ALS. AIMS MOLECULAR SCIENCE 2016. [DOI: 10.3934/molsci.2016.2.246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
675
|
Antisense Oligonucleotides for Amyotrophic Lateral Sclerosis. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
676
|
Esanov R, Belle KC, van Blitterswijk M, Belzil VV, Rademakers R, Dickson DW, Petrucelli L, Boylan KB, Dykxhoorn DM, Wuu J, Benatar M, Wahlestedt C, Zeier Z. C9orf72 promoter hypermethylation is reduced while hydroxymethylation is acquired during reprogramming of ALS patient cells. Exp Neurol 2015; 277:171-177. [PMID: 26746986 DOI: 10.1016/j.expneurol.2015.12.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/17/2015] [Accepted: 12/29/2015] [Indexed: 12/13/2022]
Abstract
Among several genetic mutations known to cause amyotrophic lateral sclerosis (ALS), a hexanucleotide repeat expansion in the C9orf72 gene is the most common. In approximately 30% of C9orf72-ALS cases, 5-methylcytosine (5mC) levels within the C9orf72 promoter are increased, resulting in a modestly attenuated phenotype. The developmental timing of C9orf72 promoter hypermethylation and the reason why it occurs in only a subset of patients remain unknown. In order to model the acquisition of C9orf72 hypermethylation and examine the potential role of 5-hydroxymethylcytosine (5hmC), we generated induced pluripotent stem cells (iPSCs) from an ALS patient with C9orf72 promoter hypermethylation. Our data show that 5mC levels are reduced by reprogramming and then re-acquired upon neuronal specification, while 5hmC levels increase following reprogramming and are highest in iPSCs and motor neurons. We confirmed the presence of 5hmC within the C9orf72 promoter in post-mortem brain tissues of hypermethylated patients. These findings show that iPSCs are a valuable model system for examining epigenetic perturbations caused by the C9orf72 mutation and reveal a potential role for cytosine demethylation.
Collapse
Affiliation(s)
- Rustam Esanov
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kinsley C Belle
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, USA; The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, USA
| | | | | | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Kevin B Boylan
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, USA; The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, USA
| | - Joanne Wuu
- Department of Neurology, University of Miami Miller School of Medicine, USA
| | - Michael Benatar
- Department of Neurology, University of Miami Miller School of Medicine, USA
| | - Claes Wahlestedt
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Zane Zeier
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
677
|
Differential Toxicity of Nuclear RNA Foci versus Dipeptide Repeat Proteins in a Drosophila Model of C9ORF72 FTD/ALS. Neuron 2015; 87:1207-1214. [PMID: 26402604 DOI: 10.1016/j.neuron.2015.09.015] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/24/2015] [Accepted: 09/09/2015] [Indexed: 12/14/2022]
Abstract
Dipeptide repeat (DPR) proteins are toxic in various models of FTD/ALS with GGGGCC (G4C2) repeat expansion. However, it is unclear whether nuclear G4C2 RNA foci also induce neurotoxicity. Here, we describe a Drosophila model expressing 160 G4C2 repeats (160R) flanked by human intronic and exonic sequences. Spliced intronic 160R formed nuclear G4C2 sense RNA foci in glia and neurons about ten times more abundantly than in human neurons; however, they had little effect on global RNA processing and neuronal survival. In contrast, highly toxic 36R in the context of poly(A)(+) mRNA were exported to the cytoplasm, where DPR proteins were produced at >100-fold higher level than in 160R flies. Moreover, the modest toxicity of intronic 160R expressed at higher temperature correlated with increased DPR production, but not RNA foci. Thus, nuclear RNA foci are neutral intermediates or possibly neuroprotective through preventing G4C2 RNA export and subsequent DPR production.
Collapse
|
678
|
Benatar M, Boylan K, Jeromin A, Rutkove SB, Berry J, Atassi N, Bruijn L. ALS biomarkers for therapy development: State of the field and future directions. Muscle Nerve 2015; 53:169-82. [PMID: 26574709 DOI: 10.1002/mus.24979] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2015] [Indexed: 12/11/2022]
Abstract
Biomarkers have become the focus of intense research in the field of amyotrophic lateral sclerosis (ALS), with the hope that they might aid therapy development efforts. Notwithstanding the discovery of many candidate biomarkers, none have yet emerged as validated tools for drug development. In this review we present a nuanced view of biomarkers based on the perspective of the Food and Drug Administration; highlight the distinction between discovery and validation; describe existing and emerging resources; review leading biological fluid-based, electrophysiological, and neuroimaging candidates relevant to therapy development efforts; discuss lessons learned from biomarker initiatives in related neurodegenerative diseases; and outline specific steps that we, as a field, might take to hasten the development and validation of biomarkers that will prove useful in enhancing efforts to develop effective treatments for ALS patients. Most important among these is the proposal to establish a federated ALS Biomarker Consortium in which all interested and willing stakeholders may participate with equal opportunity to contribute to the broader mission of biomarker development and validation.
Collapse
Affiliation(s)
- Michael Benatar
- Department of Neurology, University of Miami, Miami, Florida, USA, 33136
| | - Kevin Boylan
- Department of Neurology, Mayo Clinic Jacksonville, Jacksonville, Florida, USA
| | | | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - James Berry
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nazem Atassi
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
679
|
Methylation of C9orf72 expansion reduces RNA foci formation and dipeptide-repeat proteins expression in cells. Neurosci Lett 2015; 612:204-209. [PMID: 26690922 DOI: 10.1016/j.neulet.2015.12.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 12/13/2022]
Abstract
A hexanucleotide repeat expansion in the C9orf72 gene is the most common genetic cause of both frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), together referred to as c9FTD/ALS. It has been suggested that a loss of C9orf72 protein expression, the formation of toxic RNA foci and dipeptide-repeat proteins contribute to C9orf72-related diseases. Interestingly, it has been shown that trimethylation of histones and methylation of CpG islands near the repeat expansion may play a role in the pathogenesis c9FTD/ALS. Recently, methylation of expanded repeat itself has been reported. To further elucidate the mechanisms underlying these diseases, the influence of epigenetic modification in the repeat expansion on its pathogenic effect was assessed. Here, a reduced formation of toxic RNA foci and dipeptide-repeat proteins upon methylation of the GGGGCC repeat in a cellular model of c9FTD/ALS is shown. Additionally, a novel methylcytosine-capture DNA hybridization immunoassay for semi-quantitative detection of the repeat methylation levels is presented, potentially usable for methylation analysis in patients carrying C9orf72 repeat expansion carriers as a diagnostic tool. Presented results suggest that increased level of pathogenic GGGGCC expansion methylation may be sufficient to alleviate the molecular pathology of the C9orf72-related diseases.
Collapse
|
680
|
Burguete AS, Almeida S, Gao FB, Kalb R, Akins MR, Bonini NM. GGGGCC microsatellite RNA is neuritically localized, induces branching defects, and perturbs transport granule function. eLife 2015; 4:e08881. [PMID: 26650351 PMCID: PMC4758954 DOI: 10.7554/elife.08881] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 11/30/2015] [Indexed: 12/14/2022] Open
Abstract
Microsatellite expansions are the leading cause of numerous neurodegenerative disorders. Here we demonstrate that GGGGCC and CAG microsatellite repeat RNAs associated with C9orf72 in amyotrophic lateral sclerosis/frontotemporal dementia and with polyglutamine diseases, respectively, localize to neuritic granules that undergo active transport into distal neuritic segments. In cultured mammalian spinal cord neurons, the presence of neuritic GGGGCC repeat RNA correlates with neuronal branching defects, and the repeat RNA localizes to granules that label with fragile X mental retardation protein (FMRP), a transport granule component. Using a Drosophila GGGGCC expansion disease model, we characterize dendritic branching defects that are modulated by FMRP and Orb2. The human orthologs of these modifiers are misregulated in induced pluripotent stem cell-differentiated neurons (iPSNs) from GGGGCC expansion carriers. These data suggest that expanded repeat RNAs interact with the messenger RNA transport and translation machinery, causing transport granule dysfunction. This could be a novel mechanism contributing to the neuronal defects associated with C9orf72 and other microsatellite expansion diseases.
Collapse
Affiliation(s)
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, Worcester, United States
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, United States
| | - Robert Kalb
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, United States
| | - Michael R Akins
- Department of Biology, Drexel University, Philadelphia, United States
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
681
|
Davidson Y, Robinson AC, Liu X, Wu D, Troakes C, Rollinson S, Masuda-Suzukake M, Suzuki G, Nonaka T, Shi J, Tian J, Hamdalla H, Ealing J, Richardson A, Jones M, Pickering-Brown S, Snowden JS, Hasegawa M, Mann DMA. Neurodegeneration in frontotemporal lobar degeneration and motor neurone disease associated with expansions in C9orf72 is linked to TDP-43 pathology and not associated with aggregated forms of dipeptide repeat proteins. Neuropathol Appl Neurobiol 2015; 42:242-54. [PMID: 26538301 PMCID: PMC4832296 DOI: 10.1111/nan.12292] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 12/13/2022]
Abstract
Aims A hexanucleotide expansion in C9orf72 is the major genetic cause of inherited behavioural variant Frontotemporal dementia (bvFTD) and motor neurone disease (MND), although the pathological mechanism(s) underlying disease remains uncertain. Methods Using antibodies to poly‐GA, poly‐GP, poly‐GR, poly‐AP and poly‐PR proteins, we examined sections of cerebral cortex, hippocampus, thalamus, cerebellum and spinal cord, from 20 patients with bvFTD and/or MND bearing an expansion in C9orf72 for aggregated deposits of dipeptide repeat proteins (DPR). Results Antibodies to poly‐GA, poly‐GP and poly‐GR detected numerous rounded cytoplasmic inclusions (NCI) within granule cells of hippocampal dentate gyrus and those of the cerebellum, as well as ‘star‐burst’ shaped NCI in pyramidal neurones of CA3/4 region of hippocampus. NCI were uncommon in Purkinje cells, and only very rarely seen in anterior horn cells. Poly‐PA antibody detected occasional NCI within CA3/4 neurones alone, whereas poly‐PR antibody did not identify any NCI but immunostained the nucleus of anterior horn cells, CA3/4 neurones and Purkinje cells, in patients with or without expansion in C9orf72, as well as in normal controls. Poly‐GA antibody generally detected more DPR than poly‐GP, which in turn was greater than poly‐GR. All patients with bvFTD + MND or MND showed plentiful p62/TDP‐43 positive inclusions in remaining anterior horn cells. Conclusion Degeneration and loss of anterior horn cells associated with expansions in C9orf72 occurs in the absence of DPR, and implies that changes involving loss of nuclear staining for and a cytoplasmic aggregation of TDP‐43 are more likely to be the cause of this.
Collapse
Affiliation(s)
- Y Davidson
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, UK
| | - A C Robinson
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, UK
| | - X Liu
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, UK.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - D Wu
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, UK.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - C Troakes
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - S Rollinson
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - M Masuda-Suzukake
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - G Suzuki
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - T Nonaka
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - J Shi
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - J Tian
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Beijing, China
| | - H Hamdalla
- Manchester MND Care Centre, Salford Royal Hospital, Manchester, UK
| | - J Ealing
- Manchester MND Care Centre, Salford Royal Hospital, Manchester, UK
| | - A Richardson
- Cerebral Function Unit, Salford Royal Hospital, Manchester, UK
| | - M Jones
- Cerebral Function Unit, Salford Royal Hospital, Manchester, UK
| | - S Pickering-Brown
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Manchester, UK
| | - J S Snowden
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, UK.,Cerebral Function Unit, Salford Royal Hospital, Manchester, UK
| | - M Hasegawa
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - D M A Mann
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, UK
| |
Collapse
|
682
|
Peters OM, Cabrera GT, Tran H, Gendron TF, McKeon JE, Metterville J, Weiss A, Wightman N, Salameh J, Kim J, Sun H, Boylan KB, Dickson D, Kennedy Z, Lin Z, Zhang YJ, Daughrity L, Jung C, Gao FB, Sapp PC, Horvitz HR, Bosco DA, Brown SP, de Jong P, Petrucelli L, Mueller C, Brown RH. Human C9ORF72 Hexanucleotide Expansion Reproduces RNA Foci and Dipeptide Repeat Proteins but Not Neurodegeneration in BAC Transgenic Mice. Neuron 2015; 88:902-909. [PMID: 26637797 PMCID: PMC4828340 DOI: 10.1016/j.neuron.2015.11.018] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 10/14/2015] [Accepted: 11/13/2015] [Indexed: 12/12/2022]
Abstract
A non-coding hexanucleotide repeat expansion in the C9ORF72 gene is the most common mutation associated with familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). To investigate the pathological role of C9ORF72 in these diseases, we generated a line of mice carrying a bacterial artificial chromosome containing exons 1 to 6 of the human C9ORF72 gene with approximately 500 repeats of the GGGGCC motif. The mice showed no overt behavioral phenotype but recapitulated distinctive histopathological features of C9ORF72 ALS/FTD, including sense and antisense intranuclear RNA foci and poly(glycine-proline) dipeptide repeat proteins. Finally, using an artificial microRNA that targets human C9ORF72 in cultures of primary cortical neurons from the C9BAC mice, we have attenuated expression of the C9BAC transgene and the poly(GP) dipeptides. The C9ORF72 BAC transgenic mice will be a valuable tool in the study of ALS/FTD pathobiology and therapy.
Collapse
Affiliation(s)
- Owen M Peters
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Gabriela Toro Cabrera
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
- Department of Pediatrics and Gene Therapy Center University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Helene Tran
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Jeanne E McKeon
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jake Metterville
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Alexandra Weiss
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Nicholas Wightman
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Johnny Salameh
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Juyhun Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Huaming Sun
- Department of Pediatrics and Gene Therapy Center University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Kevin B Boylan
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, Florida 32224, USA
| | - Dennis Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Zack Kennedy
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Ziqiang Lin
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Lillian Daughrity
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Chris Jung
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr Way, Oakland, California 94609
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Peter C Sapp
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
- Department of Biology, and McGovern Institute for Brain Research, 77 Massachusetts Avenue, Massachusetts Institute of Technology, Cambridge, MA 02139, H.R.H. is an Investigator of the Howard Hughes Medical Institute
| | - H Robert Horvitz
- Department of Biology, and McGovern Institute for Brain Research, 77 Massachusetts Avenue, Massachusetts Institute of Technology, Cambridge, MA 02139, H.R.H. is an Investigator of the Howard Hughes Medical Institute
| | - Daryl A Bosco
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Solange P Brown
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Pieter de Jong
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr Way, Oakland, California 94609
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA
| | - Chris Mueller
- Department of Pediatrics and Gene Therapy Center University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
683
|
Wang J, Haeusler AR, Simko EAJ. Emerging role of RNA•DNA hybrids in C9orf72-linked neurodegeneration. Cell Cycle 2015; 14:526-32. [PMID: 25590632 DOI: 10.1080/15384101.2014.995490] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
RNA plays an active role in structural polymorphism of the genome through the formation of stable RNA•DNA hybrids (R-loops). R-loops can modulate normal physiological processes and are also associated with pathological conditions, such as those related to nucleotide repeat expansions. A guanine-rich hexanucleotide repeat expansion in chromosome 9 open reading frame 72 (C9orf72) has been linked to a spectrum of neurological conditions including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we discuss the possible roles, both locally and genome-wide, of R-loops that may arise from the C9orf72 hexanucleotide repeat. R-loops have the potential to influence the pathological processes identified in many repeat expansion diseases, such as repeat instability, transcriptional dysregulation, epigenetic modification, and antisense-mediated gene regulation. We propose that, given the wide-ranging consequences of R-loops in the cell, these structures could underlie multiple pathological processes in C9orf72-linked neurodegeneration.
Collapse
Affiliation(s)
- Jiou Wang
- a Department of Biochemistry and Molecular Biology; Bloomberg School of Public Health ; Johns Hopkins University ; Baltimore , MD USA
| | | | | |
Collapse
|
684
|
Quantitative analysis and clinico-pathological correlations of different dipeptide repeat protein pathologies in C9ORF72 mutation carriers. Acta Neuropathol 2015; 130:845-61. [PMID: 26374446 DOI: 10.1007/s00401-015-1476-2] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 09/01/2015] [Accepted: 09/03/2015] [Indexed: 12/13/2022]
Abstract
Hexanucleotide repeat expansion in C9ORF72 is the most common genetic cause of frontotemporal dementia and motor neuron disease. One consequence of the mutation is the formation of different potentially toxic polypeptides composed of dipeptide repeats (DPR) (poly-GA, -GP, -GR, -PA, -PR) generated by repeat-associated non-ATG (RAN) translation. While previous studies focusing on poly-GA pathology have failed to detect any clinico-pathological correlations in C9ORF72 mutation cases, recent data from animal and cell culture models suggested that it may be only specific DPR species that are toxic and only when accumulated in certain intracellular compartments. Therefore, we performed a systematic clinico-pathological correlative analysis with counting of actual numbers of distinct types of inclusion (neuronal cytoplasmic and intranuclear inclusions, dystrophic neurites) for each DPR protein in relevant brain regions (premotor cortex, lower motor neurons) in a cohort of 35 C9ORF72 mutation cases covering the clinical spectrum from those with pure MND, mixed FTD/MND and pure FTD. While each DPR protein pathology had a similar pattern of anatomical distribution, the total amount of inclusions for each DPR protein varied remarkably (poly-GA > GP > GR > PR/PA), indicating that RAN translation seems to be more effective from sense than from antisense transcripts. Importantly, with the exception of moderate associations for the amount of poly-GA-positive dystrophic neurites with degeneration in the frontal cortex and total burden of poly-GA pathology with disease onset, no relationship was identified for any other DPR protein pathology with degeneration or phenotype. Biochemical analysis revealed a close correlation between insoluble DPR protein species and numbers of visible inclusions, while we did not find any evidence for the presence of soluble DPR protein species. Thus, overall our findings strongly argue against a role of DPR protein aggregation as major and exclusive pathomechanism in C9ORF72 pathogenesis. However, this does not exclude that DPR protein formation might be essential in C9ORF72 pathogenesis in interplay with other consequences associated with the C9ORF72 repeat expansion.
Collapse
|
685
|
van Blitterswijk M, Gendron TF, Baker MC, DeJesus-Hernandez M, Finch NA, Brown PH, Daughrity LM, Murray ME, Heckman MG, Jiang J, Lagier-Tourenne C, Edbauer D, Cleveland DW, Josephs KA, Parisi JE, Knopman DS, Petersen RC, Petrucelli L, Boeve BF, Graff-Radford NR, Boylan KB, Dickson DW, Rademakers R. Novel clinical associations with specific C9ORF72 transcripts in patients with repeat expansions in C9ORF72. Acta Neuropathol 2015; 130:863-76. [PMID: 26437865 DOI: 10.1007/s00401-015-1480-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/14/2015] [Accepted: 09/18/2015] [Indexed: 12/12/2022]
Abstract
The loss of chromosome 9 open reading frame 72 (C9ORF72) expression, associated with C9ORF72 repeat expansions, has not been examined systematically. Three C9ORF72 transcript variants have been described thus far; the GGGGCC repeat is located between two non-coding exons (exon 1a and exon 1b) in the promoter region of transcript variant 2 (NM_018325.4) or in the first intron of variant 1 (NM_145005.6) and variant 3 (NM_001256054.2). We studied C9ORF72 expression in expansion carriers (n = 56) for whom cerebellum and/or frontal cortex was available. Using quantitative real-time PCR and digital molecular barcoding techniques, we assessed total C9ORF72 transcripts, variant 1, variant 2, variant 3, and intron containing transcripts [upstream of the expansion (intron 1a) and downstream of the expansion (intron 1b)]; the latter were correlated with levels of poly(GP) and poly(GA) proteins aberrantly translated from the expansion as measured by immunoassay (n = 50). We detected a decrease in expansion carriers as compared to controls for total C9ORF72 transcripts, variant 1, and variant 2: the strongest association was observed for variant 2 (quantitative real-time PCR cerebellum: median 43 %, p = 1.26e-06, and frontal cortex: median 58 %, p = 1.11e-05; digital molecular barcoding cerebellum: median 31 %, p = 5.23e-10, and frontal cortex: median 53 %, p = 5.07e-10). Importantly, we revealed that variant 1 levels greater than the 25th percentile conferred a survival advantage [digital molecular barcoding cerebellum: hazard ratio (HR) 0.31, p = 0.003, and frontal cortex: HR 0.23, p = 0.0001]. When focusing on intron containing transcripts, analysis of the frontal cortex revealed an increase of potentially truncated transcripts in expansion carriers as compared to controls [digital molecular barcoding frontal cortex (intron 1a): median 272 %, p = 0.003], with the highest levels in patients pathologically diagnosed with frontotemporal lobar degeneration. In the cerebellum, our analysis suggested that transcripts were less likely to be truncated and, excitingly, we discovered that intron containing transcripts were associated with poly(GP) levels [digital molecular barcoding cerebellum (intron 1a): r = 0.33, p = 0.02, and (intron 1b): r = 0.49, p = 0.0004] and poly(GA) levels [digital molecular barcoding cerebellum (intron 1a): r = 0.34, p = 0.02, and (intron 1b): r = 0.38, p = 0.007]. In summary, we report decreased expression of specific C9ORF72 transcripts and provide support for the presence of truncated transcripts as well as pre-mRNAs that may serve as templates for RAN translation. We further show that higher C9ORF72 levels may have beneficial effects, which warrants caution in the development of new therapeutic approaches.
Collapse
|
686
|
Gijselinck I, Van Mossevelde S, van der Zee J, Sieben A, Philtjens S, Heeman B, Engelborghs S, Vandenbulcke M, De Baets G, Bäumer V, Cuijt I, Van den Broeck M, Peeters K, Mattheijssens M, Rousseau F, Vandenberghe R, De Jonghe P, Cras P, De Deyn PP, Martin JJ, Cruts M, Van Broeckhoven C. Loss of TBK1 is a frequent cause of frontotemporal dementia in a Belgian cohort. Neurology 2015; 85:2116-25. [PMID: 26581300 PMCID: PMC4691687 DOI: 10.1212/wnl.0000000000002220] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/18/2015] [Indexed: 11/15/2022] Open
Abstract
Objective: To assess the genetic contribution of TBK1, a gene implicated in amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and FTD-ALS, in Belgian FTD and ALS patient cohorts containing a significant part of genetically unresolved patients. Methods: We sequenced TBK1 in a hospital-based cohort of 482 unrelated patients with FTD and FTD-ALS and 147 patients with ALS and an extended Belgian FTD-ALS family DR158. We followed up mutation carriers by segregation studies, transcript and protein expression analysis, and immunohistochemistry. Results: We identified 11 patients carrying a loss-of-function (LOF) mutation resulting in an overall mutation frequency of 1.7% (11/629), 1.1% in patients with FTD (5/460), 3.4% in patients with ALS (5/147), and 4.5% in patients with FTD-ALS (1/22). We found 1 LOF mutation, p.Glu643del, in 6 unrelated patients segregating with disease in family DR158. Of 2 mutation carriers, brain and spinal cord was characterized by TDP-43-positive pathology. The LOF mutations including the p.Glu643del mutation led to loss of transcript or protein in blood and brain. Conclusions: TBK1 LOF mutations are the third most frequent cause of clinical FTD in the Belgian clinically based patient cohort, after C9orf72 and GRN, and the second most common cause of clinical ALS after C9orf72. These findings reinforce that FTD and ALS belong to the same disease continuum.
Collapse
Affiliation(s)
- Ilse Gijselinck
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Sara Van Mossevelde
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Julie van der Zee
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Anne Sieben
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Stéphanie Philtjens
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Bavo Heeman
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Sebastiaan Engelborghs
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Mathieu Vandenbulcke
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Greet De Baets
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Veerle Bäumer
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Ivy Cuijt
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Marleen Van den Broeck
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Karin Peeters
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Maria Mattheijssens
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Frederic Rousseau
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Rik Vandenberghe
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Peter De Jonghe
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Patrick Cras
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Peter P De Deyn
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Jean-Jacques Martin
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands
| | - Marc Cruts
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands.
| | - Christine Van Broeckhoven
- From the Department of Molecular Genetics (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., M.C., C.V.B.), VIB, Antwerp; Institute Born-Bunge (I.G., S.V.M., J.v.d.Z., A.S., S.P., B.H., S.E., V.B., I.C., M.V.d.B., K.P., M.M., P.D.J., P.C., P.P.D.D., J.-J.M., M.C., C.V.B.), University of Antwerp; the Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; the Department of Neurology and Memory Clinic (S.E., P.P.D.D.), Hospital Network Antwerp Middelheim and Hoge Beuken; the Brain and Emotion Laboratory, Department of Psychiatry (M.V.), SWITCH Laboratory, VIB (G.D.B., F.R.), and Laboratory for Cognitive Neurology, Department of Neurology (R.V.), University of Leuven; the Department of Neurology (M.V., R.V.), University Hospitals Leuven, Gasthuisberg; and the Department of Neurology (P.D.J., P.C.), Antwerp University Hospital, Edegem, Belgium. P.P.D.D. is also affiliated with the Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, the Netherlands.
| | | |
Collapse
|
687
|
Zhou B, Liu C, Geng Y, Zhu G. Topology of a G-quadruplex DNA formed by C9orf72 hexanucleotide repeats associated with ALS and FTD. Sci Rep 2015; 5:16673. [PMID: 26564809 PMCID: PMC4643247 DOI: 10.1038/srep16673] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/07/2015] [Indexed: 12/13/2022] Open
Abstract
Abnormal expansions of an intronic hexanucleotide GGGGCC (G4C2) repeat of the C9orf72 gene are the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Previous studies suggested that the C9orf72 hexanucleotide repeat expansion (HRE), either as DNA or the transcribed RNA, can fold into G-quadruplexes with distinct structures. These structural polymorphisms lead to abortive transcripts and contribute to the pathogenesis of ALS and FTD. Using circular dichroism (CD) and nuclear magnetic resonance (NMR) spectroscopy, we analyzed the structures of C9orf72 HRE DNA with various G4C2 repeats. They exhibited diverse G-quadruplex folds in potassium ions. Furthermore, we determined the topology of a G-quadruplex formed by d(G4C2)4. It favors a monomeric fold and forms a chair-type G-quadruplex with a four-layer antiparallel G-tetra core and three edgewise loops, which is distinct from known structures of chair-type G-quadruplexes. Our findings highlight the conformational heterogeneity of C9orf72 HRE DNA, and may lay the necessary structural basis for designing small molecules for the modulation of ALS/FTD pathogenesis.
Collapse
Affiliation(s)
- Bo Zhou
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, PRC
| | - Changdong Liu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, PRC
| | - Yanyan Geng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, PRC
| | - Guang Zhu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, PRC
| |
Collapse
|
688
|
Buijsen RAM, Visser JA, Kramer P, Severijnen EAWFM, Gearing M, Charlet-Berguerand N, Sherman SL, Berman RF, Willemsen R, Hukema RK. Presence of inclusions positive for polyglycine containing protein, FMRpolyG, indicates that repeat-associated non-AUG translation plays a role in fragile X-associated primary ovarian insufficiency. Hum Reprod 2015; 31:158-68. [PMID: 26537920 DOI: 10.1093/humrep/dev280] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/15/2015] [Indexed: 12/11/2022] Open
Abstract
STUDY QUESTION Does repeat-associated non-AUG (RAN) translation play a role in fragile X-associated primary ovarian insufficiency (FXPOI), leading to the presence of polyglycine containing protein (FMRpolyG)-positive inclusions in ovarian tissue? SUMMARY ANSWER Ovaries of a woman with FXPOI and of an Fmr1 premutation (PM) mouse model (exCGG-KI) contain intranuclear inclusions that stain positive for both FMRpolyG and ubiquitin. WHAT IS KNOWN ALREADY Women who carry the FMR1 PM are at 20-fold increased risk to develop primary ovarian insufficiency (FXPOI). A toxic RNA gain-of-function has been suggested as the underlying mechanism since the PM results in increased levels of mRNA containing an expanded repeat, but reduced protein levels of fragile X mental retardation protein (FMRP). Recently, RAN translation has been shown to occur from FMR1 mRNA that contains PM repeat expansions, leading to FMRpolyG inclusions in brain and non-CNS tissues of fragile X-associated tremor/ataxia syndrome (FXTAS) patients. STUDY DESIGN, SIZE, DURATION Ovaries of a woman with FXPOI and women without PM (controls), and ovaries from wild-type and exCGG-KI mice were analyzed by immunohistochemistry for the presence of inclusions that stained for ubiquitin and FMRpolyG . The ovaries from wild-type and exCGG-KI mice were further characterized for the number of follicles, Fmr1 mRNA levels and FMRP protein expression. The presence of inclusions was also analyzed in pituitaries of a man with FXTAS and the exCGG-KI mice. PARTICIPANTS/MATERIALS, SETTING, METHODS Human ovaries from a woman with FXPOI and two control subjects and pituitaries from a man with FXTAS and a control subjects were fixed in 4% formalin. Ovaries and pituitaries of wild-type and exCGG mice were fixed in Bouin's fluid or 4% paraformaldehyde. Immunohistochemistry was performed on the human and mouse samples using FMRpolyG, ubiquitin and Fmrp antibodies. Fmr1 mRNA and protein expression were determined in mouse ovaries by quantitative RT-PCR and Western blot analysis. Follicle numbers in mouse ovaries were determined in serial sections by microscopy. MAIN RESULTS AND THE ROLE OF CHANCE FMRpolyG-positive inclusions were present in ovarian stromal cells of a woman with FXPOI but not in the ovaries of control subjects. The FMRpolyG-positive inclusions colocalized with ubiquitin-positive inclusions. Similar inclusions were also observed in the pituitary of a man with FXTAS but not in control subjects. Similarly, ovaries of 40-week-old exCGG-KI mice, but not wild-type mice, contained numerous inclusions in the stromal cells that stained for both FMRpolyG- and ubiquitin, while the ovaries of 20-week-old exCGG-KI contained fewer inclusions. At 40 weeks ovarian Fmr1 mRNA expression was increased by 5-fold in exCGG-KI mice compared with wild-type mice, while Fmrp expression was reduced by 2-fold. With respect to ovarian function in exCGG-KI mice: (i) although the number of healthy growing follicles did not differ between wild-type and exCGG-KI mice, the number of atretic large antral follicles was increased by nearly 9-fold in 40-week old exCGG-KI mice (P < 0.001); (ii) at 40 weeks of age only 50% of exCGG-KI mice had recent ovulations compared with 89% in wild-type mice (P = 0.07) and (iii) those exCGG-KI mice with recent ovulations tended to have a reduced number of fresh corpora lutea (4.8 ± 1.74 versus 8.50 ± 0.98, exCGG-KI versus wild-type mice, respectively, P = 0.07). LIMITATIONS, REASONS FOR CAUTION Although FMRpolyG-positive inclusions were detected in ovaries of both a woman with FXPOI and a mouse model of the FMR1 PM, we only analyzed one ovary from a FXPOI subject. Caution is needed to extrapolate these results to all women with the FMR1 PM. Furthermore, the functional consequence of FMRpolyG-positive inclusions in the ovaries for reproduction remains to be determined. WIDER IMPLICATIONS OF THE FINDINGS Our results suggest that a dysfunctional hypothalamic-pituitary-gonadal-axis may contribute to FXPOI in FMR1 PM carriers. STUDY FUNDING/COMPETING INTERESTS This study was supported by grants from NFXF, ZonMW, the Netherlands Brain Foundation and NIH. The authors have no conflict of interest to declare.
Collapse
Affiliation(s)
- R A M Buijsen
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - J A Visser
- Department of Internal Medicine, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - P Kramer
- Department of Internal Medicine, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - E A W F M Severijnen
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - M Gearing
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - N Charlet-Berguerand
- Department of Neurobiology and Genetics, IGBMC, INSERM U964, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - S L Sherman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - R F Berman
- Department of Neurological Surgery, UC Davis, Davis 95618, CA, USA
| | - R Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| | - R K Hukema
- Department of Clinical Genetics, Erasmus Medical Center, 3015 CE Rotterdam, The Netherlands
| |
Collapse
|
689
|
Wallon D, Nicolas G. Genetica delle demenze degenerative. Neurologia 2015. [DOI: 10.1016/s1634-7072(15)73962-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
690
|
Gozes I, Ivashko-Pachima Y. ADNP: in search for molecular mechanisms and innovative therapeutic strategies for frontotemporal degeneration. Front Aging Neurosci 2015; 7:205. [PMID: 26578950 PMCID: PMC4624849 DOI: 10.3389/fnagi.2015.00205] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/12/2015] [Indexed: 12/04/2022] Open
Abstract
Activity-dependent neuroprotective protein (ADNP) is deregulated in Alzheimer's disease (AD) and in schizophrenia and mutated in autism. In mice, ADNP is essential for brain formation and ADNP haploinsufficiency is associated with cognitive and social deficits and tauopathy. Tauopathy, a major pathology in AD, is also found in ~45% of frontotemporal dementias (FTDs). Tau transcript, a product of a single gene, undergoes alternative splicing. Tau splicing seems to be altered in FTD brain. In transgenic mice overexpressing a mutated tau in the cerebral cortex, significant increases in ADNP transcript expression were observed in the cerebral cortex of young transgenic mice (~disease onset) and a marked decrease with aging as compared to control littermates. ADNP is a member of the SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeling complex also associated with alternative splicing, including tau transcript splicing. Further cellular interactions of ADNP include association with microtubules, with tau being a microtubule—associated protein. NAP (davundetide), a novel drug candidate derived from ADNP, increases ADNP-microtubule association and protects against tauopathy and cognitive deficiencies in mice. Although, NAP did not provide protection in progressive supranuclear palsy (PSP), a pure tauopathy, it increased cognitive scores in amnestic mild cognitively impaired patients and protected functional activity in schizophrenia patients. This mini-review focuses on ADNP in the context of FTD and tau/microtubules and proposes NAP as a novel drug target for future clinical evaluations.
Collapse
Affiliation(s)
- Illana Gozes
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Sagol School of Neuroscience & Adams Super Center for Brain Studies, Tel Aviv University Tel Aviv, Israel
| | - Yanina Ivashko-Pachima
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Sagol School of Neuroscience & Adams Super Center for Brain Studies, Tel Aviv University Tel Aviv, Israel
| |
Collapse
|
691
|
Childs-Disney JL, Disney MD. Approaches to Validate and Manipulate RNA Targets with Small Molecules in Cells. Annu Rev Pharmacol Toxicol 2015; 56:123-40. [PMID: 26514201 DOI: 10.1146/annurev-pharmtox-010715-103910] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RNA has become an increasingly important target for therapeutic interventions and for chemical probes that dissect and manipulate its cellular function. Emerging targets include human RNAs that have been shown to directly cause cancer, metabolic disorders, and genetic disease. In this review, we describe various routes to obtain bioactive compounds that target RNA, with a particular emphasis on the development of small molecules. We use these cases to describe approaches that are being developed for target validation, which include target-directed cleavage, classic pull-down experiments, and covalent cross-linking. Thus, tools are available to design small molecules to target RNA and to identify the cellular RNAs that are their targets.
Collapse
Affiliation(s)
| | - Matthew D Disney
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458; ,
| |
Collapse
|
692
|
Biasiotto G, Di Lorenzo D, Archetti S, Zanella I. Iron and Neurodegeneration: Is Ferritinophagy the Link? Mol Neurobiol 2015; 53:5542-74. [PMID: 26468157 DOI: 10.1007/s12035-015-9473-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/01/2015] [Indexed: 12/12/2022]
Abstract
Mounting evidence indicates that the lysosome-autophagy pathway plays a critical role in iron release from ferritin, the main iron storage cellular protein, hence in the distribution of iron to the cells. The recent identification of nuclear receptor co-activator 4 as the receptor for ferritin delivery to selective autophagy sheds further light on the understanding of the mechanisms underlying this pathway. The emerging view is that iron release from ferritin through the lysosomes is a general mechanism in normal and tumour cells of different tissue origins, but it has not yet been investigated in brain cells. Defects in the lysosome-autophagy pathway are often involved in the pathogenesis of neurodegenerative disorders, and brain iron homeostasis disruption is a hallmark of many of these diseases. However, in most cases, it has not been established whether iron dysregulation is directly involved in the pathogenesis of the diseases or if it is a secondary effect derived from other pathogenic mechanisms. The recent evidence of the crucial involvement of autophagy in cellular iron handling offers new perspectives about the role of iron in neurodegeneration, suggesting that autophagy dysregulation could cause iron dyshomeostasis. In this review, we recapitulate our current knowledge on the routes through which iron is released from ferritin, focusing on the most recent advances. We summarise the current evidence concerning lysosome-autophagy pathway dysfunctions and those of iron metabolism and discuss their potential interconnections in several neurodegenerative disorders, such as Alzheimer's, Parkinson's and Huntington's diseases; amyotrophic lateral sclerosis; and frontotemporal lobar dementia.
Collapse
Affiliation(s)
- Giorgio Biasiotto
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy
- Biotechnology Laboratory, Department of Diagnostics, Civic Hospital of Brescia, Piazzale Spedali Civili 1, 25123, Brescia, Italy
| | - Diego Di Lorenzo
- Biotechnology Laboratory, Department of Diagnostics, Civic Hospital of Brescia, Piazzale Spedali Civili 1, 25123, Brescia, Italy
| | - Silvana Archetti
- Biotechnology Laboratory, Department of Diagnostics, Civic Hospital of Brescia, Piazzale Spedali Civili 1, 25123, Brescia, Italy
| | - Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
- Biotechnology Laboratory, Department of Diagnostics, Civic Hospital of Brescia, Piazzale Spedali Civili 1, 25123, Brescia, Italy.
| |
Collapse
|
693
|
Lee S, Huang EJ. Modeling ALS and FTD with iPSC-derived neurons. Brain Res 2015; 1656:88-97. [PMID: 26462653 DOI: 10.1016/j.brainres.2015.10.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 08/05/2015] [Accepted: 10/02/2015] [Indexed: 12/14/2022]
Abstract
Recent advances in genetics and neuropathology support the idea that amyotrophic lateral sclerosis (ALS) and frontotemporal lobar dementia (FTD) are two ends of a disease spectrum. Although several animal models have been developed to investigate the pathogenesis and disease progression in ALS and FTD, there are significant limitations that hamper our ability to connect these models with the neurodegenerative processes in human diseases. With the technical breakthrough in reprogramming biology, it is now possible to generate patient-specific induced pluripotent stem cells (iPSCs) and disease-relevant neuron subtypes. This review provides a comprehensive summary of studies that use iPSC-derived neurons to model ALS and FTD. We discuss the unique capabilities of iPSC-derived neurons that capture some key features of ALS and FTD, and underscore their potential roles in drug discovery. There are, however, several critical caveats that require improvements before iPSC-derived neurons can become highly effective disease models. This article is part of a Special Issue entitled SI: Exploiting human neurons.
Collapse
Affiliation(s)
- Sebum Lee
- Department of Pathology, University of California San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143-0502, United States
| | - Eric J Huang
- Department of Pathology, University of California San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143-0502, United States; Pathology Service 113B, San Francisco VA Medical Center, 505 Parnassus Avenue, San Francisco, CA 94143-0502, United States.
| |
Collapse
|
694
|
Anor CJ, Xi Z, Zhang M, Moreno D, Sato C, Rogaeva E, Tartaglia MC. Mutation analysis of C9orf72 in patients with corticobasal syndrome. Neurobiol Aging 2015; 36:2905.e1-5. [DOI: 10.1016/j.neurobiolaging.2015.06.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/01/2015] [Accepted: 06/05/2015] [Indexed: 12/13/2022]
|
695
|
Gendron TF, van Blitterswijk M, Bieniek KF, Daughrity LM, Jiang J, Rush BK, Pedraza O, Lucas JA, Murray ME, Desaro P, Robertson A, Overstreet K, Thomas CS, Crook JE, Castanedes-Casey M, Rousseau L, Josephs KA, Parisi JE, Knopman DS, Petersen RC, Boeve BF, Graff-Radford NR, Rademakers R, Lagier-Tourenne C, Edbauer D, Cleveland DW, Dickson DW, Petrucelli L, Boylan KB. Cerebellar c9RAN proteins associate with clinical and neuropathological characteristics of C9ORF72 repeat expansion carriers. Acta Neuropathol 2015; 130:559-73. [PMID: 26350237 PMCID: PMC4575385 DOI: 10.1007/s00401-015-1474-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/29/2015] [Accepted: 08/30/2015] [Indexed: 12/12/2022]
Abstract
Clinical and neuropathological characteristics associated with G4C2 repeat expansions in chromosome 9 open reading frame 72 (C9ORF72), the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia, are highly variable. To gain insight on the molecular basis for the heterogeneity among C9ORF72 mutation carriers, we evaluated associations between features of disease and levels of two abundantly expressed “c9RAN proteins” produced by repeat-associated non-ATG (RAN) translation of the expanded repeat. For these studies, we took a departure from traditional immunohistochemical approaches and instead employed immunoassays to quantitatively measure poly(GP) and poly(GA) levels in cerebellum, frontal cortex, motor cortex, and/or hippocampus from 55 C9ORF72 mutation carriers [12 patients with ALS, 24 with frontotemporal lobar degeneration (FTLD) and 19 with FTLD with motor neuron disease (FTLD-MND)]. We additionally investigated associations between levels of poly(GP) or poly(GA) and cognitive impairment in 15 C9ORF72 ALS patients for whom neuropsychological data were available. Among the neuroanatomical regions investigated, poly(GP) levels were highest in the cerebellum. In this same region, associations between poly(GP) and both neuropathological and clinical features were detected. Specifically, cerebellar poly(GP) levels were significantly lower in patients with ALS compared to patients with FTLD or FTLD-MND. Furthermore, cerebellar poly(GP) associated with cognitive score in our cohort of 15 patients. In the cerebellum, poly(GA) levels similarly trended lower in the ALS subgroup compared to FTLD or FTLD-MND subgroups, but no association between cerebellar poly(GA) and cognitive score was detected. Both cerebellar poly(GP) and poly(GA) associated with C9ORF72 variant 3 mRNA expression, but not variant 1 expression, repeat size, disease onset, or survival after onset. Overall, these data indicate that cerebellar abnormalities, as evidenced by poly(GP) accumulation, associate with neuropathological and clinical phenotypes, in particular cognitive impairment, of C9ORF72 mutation carriers.
Collapse
Affiliation(s)
- Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Kevin F Bieniek
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Mayo Graduate School, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Jie Jiang
- Ludwig Institute, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Beth K Rush
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Otto Pedraza
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - John A Lucas
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Pamela Desaro
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Amelia Robertson
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Karen Overstreet
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Colleen S Thomas
- Section of Biostatistics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Julia E Crook
- Section of Biostatistics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Linda Rousseau
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Joseph E Parisi
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Clotilde Lagier-Tourenne
- Ludwig Institute, University of California at San Diego, La Jolla, CA, 92093, USA
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Metabolic Biochemistry, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Don W Cleveland
- Ludwig Institute, University of California at San Diego, La Jolla, CA, 92093, USA
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Kevin B Boylan
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
696
|
Distribution of dipeptide repeat proteins in cellular models and C9orf72 mutation cases suggests link to transcriptional silencing. Acta Neuropathol 2015; 130:537-55. [PMID: 26085200 PMCID: PMC4575390 DOI: 10.1007/s00401-015-1450-z] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/25/2015] [Accepted: 06/03/2015] [Indexed: 12/12/2022]
Abstract
A massive expansion of a GGGGCC repeat upstream of the C9orf72 coding region is the most common known cause of amyotrophic lateral sclerosis and frontotemporal dementia. Despite its intronic localization and lack of a canonical start codon, both strands are translated into aggregating dipeptide repeat (DPR) proteins: poly-GA, poly-GP, poly-GR, poly-PR and poly-PA. To address conflicting findings on the predominant toxicity of the different DPR species in model systems, we compared the expression pattern of the DPR proteins in rat primary neurons and postmortem brain and spinal cord of C9orf72 mutation patients. Only poly-GA overexpression closely mimicked the p62-positive neuronal cytoplasmic inclusions commonly observed for all DPR proteins in patients. In contrast, overexpressed poly-GR and poly-PR formed nucleolar p62-negative inclusions. In patients, most of the less common neuronal intranuclear DPR inclusions were para-nucleolar and p62 positive. Neuronal nucleoli in C9orf72 cases showed normal size and morphology regardless of the presence of poly-GR and poly-PR inclusions arguing against widespread nucleolar stress, reported in cellular models. Colocalization of para-nucleolar DPR inclusions with heterochromatin and a marker of transcriptional repression (H3K9me2) indicates a link to gene transcription. In contrast, we detected numerous intranuclear DPR inclusions not associated with nucleolar structures in ependymal and subependymal cells. In patients, neuronal inclusions of poly-GR, poly-GP and the poly-GA interacting protein Unc119 were less abundant than poly-GA inclusions, but showed similar regional and subcellular distribution. Regardless of neurodegeneration, all inclusions were most abundant in neocortex, hippocampus and thalamus, with few inclusions in brain stem and spinal cord. In the granular cell layer of the cerebellum, poly-GA and Unc119 inclusions were significantly more abundant in cases with FTLD than in cases with MND and FTLD/MND. Poly-PR inclusions were rare throughout the brain but significantly more abundant in the CA3/4 region of FTLD cases than in MND cases. Thus, although DPR distribution is not correlated with neurodegeneration spatially, it correlates with neuropathological subtypes.
Collapse
|
697
|
McGurk L, Berson A, Bonini NM. Drosophila as an In Vivo Model for Human Neurodegenerative Disease. Genetics 2015; 201:377-402. [PMID: 26447127 PMCID: PMC4596656 DOI: 10.1534/genetics.115.179457] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/19/2015] [Indexed: 12/13/2022] Open
Abstract
With the increase in the ageing population, neurodegenerative disease is devastating to families and poses a huge burden on society. The brain and spinal cord are extraordinarily complex: they consist of a highly organized network of neuronal and support cells that communicate in a highly specialized manner. One approach to tackling problems of such complexity is to address the scientific questions in simpler, yet analogous, systems. The fruit fly, Drosophila melanogaster, has been proven tremendously valuable as a model organism, enabling many major discoveries in neuroscientific disease research. The plethora of genetic tools available in Drosophila allows for exquisite targeted manipulation of the genome. Due to its relatively short lifespan, complex questions of brain function can be addressed more rapidly than in other model organisms, such as the mouse. Here we discuss features of the fly as a model for human neurodegenerative disease. There are many distinct fly models for a range of neurodegenerative diseases; we focus on select studies from models of polyglutamine disease and amyotrophic lateral sclerosis that illustrate the type and range of insights that can be gleaned. In discussion of these models, we underscore strengths of the fly in providing understanding into mechanisms and pathways, as a foundation for translational and therapeutic research.
Collapse
Affiliation(s)
- Leeanne McGurk
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Amit Berson
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
698
|
Lourenco GF, Janitz M, Huang Y, Halliday GM. Long noncoding RNAs in TDP-43 and FUS/TLS-related frontotemporal lobar degeneration (FTLD). Neurobiol Dis 2015. [DOI: 10.1016/j.nbd.2015.07.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
699
|
Yang D, Abdallah A, Li Z, Lu Y, Almeida S, Gao FB. FTD/ALS-associated poly(GR) protein impairs the Notch pathway and is recruited by poly(GA) into cytoplasmic inclusions. Acta Neuropathol 2015; 130:525-35. [PMID: 26031661 PMCID: PMC4575383 DOI: 10.1007/s00401-015-1448-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/16/2015] [Accepted: 05/17/2015] [Indexed: 12/14/2022]
Abstract
C9ORF72 repeat expansion is the most common genetic mutation in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Abnormal dipeptide repeat proteins (DPRs) generated from repeat-associated non-AUG (RAN) translation of repeat-containing RNAs are thought to be pathogenic; however, the mechanisms are unknown. Here we report that (GR)80 and (PR)80 are toxic in neuronal and non-neuronal cells in Drosophila. In contrast to reported shorter poly(GR) forms, (GR)80 is mostly localized throughout the cytosol without detectable accumulation in the nucleolus, accompanied by suppression of Notch signaling and cell loss in the wing. Some Notch target genes are also downregulated in brains and iPSC-derived cortical neurons of C9ORF72 patients. Increased Notch expression largely suppressed (GR)80-induced cell loss in the wing. When co-expressed in Drosophila, HeLa cells, or human neurons, (GA)80 recruited (GR)80 into cytoplasmic inclusions, partially decreasing the toxicity of (GR)80 and restoring Notch signaling in Drosophila. Thus, different DPRs have opposing roles in cell loss and we identify the Notch pathway as one of the receptor signaling pathways that might be compromised in C9ORF72 FTD/ALS.
Collapse
Affiliation(s)
- Dejun Yang
- Department of Neurology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Abbas Abdallah
- Department of Neurology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Zhaodong Li
- Department of Neurology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02216, USA
| | - Yubing Lu
- Department of Neurology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
700
|
Atkinson RAK, Fernandez-Martos CM, Atkin JD, Vickers JC, King AE. C9ORF72 expression and cellular localization over mouse development. Acta Neuropathol Commun 2015; 3:59. [PMID: 26408000 PMCID: PMC4582620 DOI: 10.1186/s40478-015-0238-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 09/15/2015] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION A majority of familial frontotemporal lobar dementia and amyotrophic lateral sclerosis cases are associated with a large repeat expansion in a non-coding region of the C9ORF72 gene. Currently, little is known about the normal function and the expression pattern of the C9ORF72 protein. The aims of this study were to characterize the expression pattern and cellular localization of the three reported mouse isoforms of C9orf72, over a developmental time-course in primary cultured cortical neurons and brain tissue from C57BL/6 mice. RESULTS We demonstrated that the different isoforms of C9ORF72 at the mRNA and protein level undergo alterations in expression during development and into adulthood. Cellular fractionation and immunofluorescence demonstrated that levels of nuclear and cytoplasmic expression of isoforms changed significantly over the time course. Additionally, immunofluorescence studies showed C9ORF72 labeling as puncta throughout neurons, extending beyond the microtubule cytoskeleton into actin-rich structures such as filopodia and growth cones. Finally, synaptosome preparations demonstrated the presence of C9ORF72 isoform 1 in synaptic-rich fractions from adult mouse brain. CONCLUSION In summary, the presence of C9ORF72 as puncta and within synaptic-rich fractions may indicate involvement at the synapse and differential expression of isoforms in nuclei and cytoplasm may suggest distinct roles for the isoforms. Determining the physiological role of C9ORF72 protein may help to determine the role it plays in disease.
Collapse
|