51
|
Bellmann J, Monette A, Tripathy V, Sójka A, Abo-Rady M, Janosh A, Bhatnagar R, Bickle M, Mouland AJ, Sterneckert J. Viral Infections Exacerbate FUS-ALS Phenotypes in iPSC-Derived Spinal Neurons in a Virus Species-Specific Manner. Front Cell Neurosci 2019; 13:480. [PMID: 31695598 PMCID: PMC6817715 DOI: 10.3389/fncel.2019.00480] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) arises from an interplay of genetic mutations and environmental factors. ssRNA viruses are possible ALS risk factors, but testing their interaction with mutations such as in FUS, which encodes an RNA-binding protein, has been difficult due to the lack of a human disease model. Here, we use isogenic induced pluripotent stem cell (iPSC)-derived spinal neurons (SNs) to investigate the interaction between ssRNA viruses and mutant FUS. We find that rabies virus (RABV) spreads ALS phenotypes, including the formation of stress granules (SGs) with aberrant composition due to increased levels of FUS protein, as well as neurodegeneration and reduced restriction activity by FUS mutations. Consistent with this, iPSC-derived SNs harboring mutant FUS are more sensitive to human immunodeficiency virus (HIV-1) and Zika viruses (ZIKV). We demonstrate that RABV and HIV-1 exacerbate cytoplasmic mislocalization of FUS. Our results demonstrate that viral infections worsen ALS pathology in SNs with genetic risk factors, suggesting a novel role for viruses in modulating patient phenotypes.
Collapse
Affiliation(s)
- Jessica Bellmann
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Anne Monette
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Vadreenath Tripathy
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Anna Sójka
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Masin Abo-Rady
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Antje Janosh
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Marc Bickle
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrew J Mouland
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Jared Sterneckert
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
52
|
Cappella M, Ciotti C, Cohen-Tannoudji M, Biferi MG. Gene Therapy for ALS-A Perspective. Int J Mol Sci 2019; 20:E4388. [PMID: 31500113 PMCID: PMC6771059 DOI: 10.3390/ijms20184388] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease (MND) with no cure. Recent advances in gene therapy open a new perspective to treat this disorder-particularly for the characterized genetic forms. Gene therapy approaches, involving the delivery of antisense oligonucleotides into the central nervous system (CNS) are being tested in clinical trials for patients with mutations in SOD1 or C9orf72 genes. Viral vectors can be used to deliver therapeutic sequences to stably transduce motor neurons in the CNS. Vectors derived from adeno-associated virus (AAV), can efficiently target genes and have been tested in several pre-clinical settings with promising outcomes. Recently, the Food and Drug Administration (FDA) approved Zolgensma, an AAV-mediated treatment for another MND-the infant form of spinal muscular atrophy. Given the accelerated progress in gene therapy, it is potentially a promising avenue to develop an efficient and safe cure for ALS.
Collapse
Affiliation(s)
- Marisa Cappella
- Sorbonne Université, Inserm UMRS 974, Centre of Research in Myology (CRM), Institut de Myologie, GH Pitié Salpêtrière, 75013 Paris, France
| | - Chiara Ciotti
- Sorbonne Université, Inserm UMRS 974, Centre of Research in Myology (CRM), Institut de Myologie, GH Pitié Salpêtrière, 75013 Paris, France
| | - Mathilde Cohen-Tannoudji
- Sorbonne Université, Inserm UMRS 974, Centre of Research in Myology (CRM), Institut de Myologie, GH Pitié Salpêtrière, 75013 Paris, France
| | - Maria Grazia Biferi
- Sorbonne Université, Inserm UMRS 974, Centre of Research in Myology (CRM), Institut de Myologie, GH Pitié Salpêtrière, 75013 Paris, France.
| |
Collapse
|
53
|
Nair RR, Corrochano S, Gasco S, Tibbit C, Thompson D, Maduro C, Ali Z, Fratta P, Arozena AA, Cunningham TJ, Fisher EMC. Uses for humanised mouse models in precision medicine for neurodegenerative disease. Mamm Genome 2019; 30:173-191. [PMID: 31203387 PMCID: PMC6759662 DOI: 10.1007/s00335-019-09807-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 05/11/2019] [Indexed: 12/11/2022]
Abstract
Neurodegenerative disease encompasses a wide range of disorders afflicting the central and peripheral nervous systems and is a major unmet biomedical need of our time. There are very limited treatments, and no cures, for most of these diseases, including Alzheimer's Disease, Parkinson's Disease, Huntington Disease, and Motor Neuron Diseases. Mouse and other animal models provide hope by analysing them to understand pathogenic mechanisms, to identify drug targets, and to develop gene therapies and stem cell therapies. However, despite many decades of research, virtually no new treatments have reached the clinic. Increasingly, it is apparent that human heterogeneity within clinically defined neurodegenerative disorders, and between patients with the same genetic mutations, significantly impacts disease presentation and, potentially, therapeutic efficacy. Therefore, stratifying patients according to genetics, lifestyle, disease presentation, ethnicity, and other parameters may hold the key to bringing effective therapies from the bench to the clinic. Here, we discuss genetic and cellular humanised mouse models, and how they help in defining the genetic and environmental parameters associated with neurodegenerative disease, and so help in developing effective precision medicine strategies for future healthcare.
Collapse
Affiliation(s)
- Remya R Nair
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Silvia Corrochano
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Samanta Gasco
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Charlotte Tibbit
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - David Thompson
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Cheryl Maduro
- Department of Neuromuscular Diseases, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Zeinab Ali
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK
| | - Pietro Fratta
- Department of Neuromuscular Diseases, Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Abraham Acevedo Arozena
- Unidad de Investigación Hospital Universitario de Canarias, FUNCANIS, Instituto de Tecnologías Biomédicas ULL, and CIBERNED, La Laguna, 38320, Tenerife, Spain
| | | | - Elizabeth M C Fisher
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, OX11 0RD, UK.
- Department of Neuromuscular Diseases, Institute of Neurology, University College London, London, WC1N 3BG, UK.
| |
Collapse
|
54
|
|
55
|
Increased FUS levels in astrocytes leads to astrocyte and microglia activation and neuronal death. Sci Rep 2019; 9:4572. [PMID: 30872738 PMCID: PMC6418113 DOI: 10.1038/s41598-019-41040-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
Mutations of Fused in sarcoma (FUS), a ribonucleoprotein involved in RNA metabolism, have been found associated with both familial and sporadic cases of amyotrophic lateral sclerosis (ALS). Notably, besides mutations in the coding sequence, also mutations into the 3′ untranslated region, leading to increased levels of the wild-type protein, have been associated with neuronal death and ALS pathology, in ALS models and patients. The mechanistic link between altered FUS levels and ALS-related neurodegeneration is far to be elucidated, as well as the consequences of elevated FUS levels in the modulation of the inflammatory response sustained by glial cells, a well-recognized player in ALS progression. Here, we studied the effect of wild-type FUS overexpression on the responsiveness of mouse and human neural progenitor-derived astrocytes to a pro-inflammatory stimulus (IL1β) used to mimic an inflammatory environment. We found that astrocytes with increased FUS levels were more sensitive to IL1β, as shown by their enhanced expression of inflammatory genes, compared with control astrocytes. Moreover, astrocytes overexpressing FUS promoted neuronal cell death and pro-inflammatory microglia activation. We conclude that overexpression of wild-type FUS intrinsically affects astrocyte reactivity and drives their properties toward pro-inflammatory and neurotoxic functions, suggesting that a non-cell autonomous mechanism can support neurodegeneration in FUS-mutated animals and patients.
Collapse
|
56
|
Larner AJ, Fratalia L. Focal limb weakness (monoparesis): when family history holds the key to diagnosis. Br J Hosp Med (Lond) 2019; 80:110-111. [DOI: 10.12968/hmed.2019.80.2.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- AJ Larner
- Consultant Neurologist, Walton Centre for Neurology and Neurosurgery, Liverpool L9 7LJ
| | - L Fratalia
- Consultant Neurologist, Walton Centre for Neurology and Neurosurgery, Liverpool
| |
Collapse
|
57
|
Nicolas G, Veltman JA. The role of de novo mutations in adult-onset neurodegenerative disorders. Acta Neuropathol 2019; 137:183-207. [PMID: 30478624 PMCID: PMC6513904 DOI: 10.1007/s00401-018-1939-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 12/13/2022]
Abstract
The genetic underpinnings of the most common adult-onset neurodegenerative disorders (AOND) are complex in majority of the cases. In some families, however, the disease can be inherited in a Mendelian fashion as an autosomal-dominant trait. Next to that, patients carrying mutations in the same disease genes have been reported despite a negative family history. Although challenging to demonstrate due to the late onset of the disease in most cases, the occurrence of de novo mutations can explain this sporadic presentation, as demonstrated for severe neurodevelopmental disorders. Exome or genome sequencing of patient-parent trios allows a hypothesis-free study of the role of de novo mutations in AOND and the discovery of novel disease genes. Another hypothesis that may explain a proportion of sporadic AOND cases is the occurrence of a de novo mutation after the fertilization of the oocyte (post-zygotic mutation) or even as a late-somatic mutation, restricted to the brain. Such somatic mutation hypothesis, that can be tested with the use of novel sequencing technologies, is fully compatible with the seeding and spreading mechanisms of the pathological proteins identified in most of these disorders. We review here the current knowledge and future perspectives on de novo mutations in known and novel candidate genes identified in the most common AONDs such as Alzheimer's disease, Parkinson's disease, the frontotemporal lobar degeneration spectrum and Prion disorders. Also, we review the first lessons learned from recent genomic studies of control and diseased brains and the challenges which remain to be addressed.
Collapse
Affiliation(s)
- Gaël Nicolas
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, 22, Boulevard Gambetta, 76000, 76031, Rouen Cedex, France.
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Joris A Veltman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
58
|
Eura N, Sugie K, Suzuki N, Kiriyama T, Izumi T, Shimakura N, Kato M, Aoki M. A juvenile sporadic amyotrophic lateral sclerosis case with P525L mutation in the FUS gene: A rare co-occurrence of autism spectrum disorder and tremor. J Neurol Sci 2019; 398:67-68. [PMID: 30684766 DOI: 10.1016/j.jns.2019.01.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Nobuyuki Eura
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Nara, Japan.
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Nara, Japan.
| | - Naoki Suzuki
- Department of Neurology, Tohoku University, Sendai, Miyagi, Japan.
| | - Takao Kiriyama
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Nara, Japan.
| | - Tesseki Izumi
- Department of Neurology, Nara Medical University School of Medicine, Kashihara, Nara, Japan.
| | - Naoko Shimakura
- Department of Neurology, Tohoku University, Sendai, Miyagi, Japan.
| | - Masaaki Kato
- Department of Neurology, Tohoku University, Sendai, Miyagi, Japan.
| | - Masashi Aoki
- Department of Neurology, Tohoku University, Sendai, Miyagi, Japan.
| |
Collapse
|
59
|
Guo L, Fare CM, Shorter J. Therapeutic Dissolution of Aberrant Phases by Nuclear-Import Receptors. Trends Cell Biol 2019; 29:308-322. [PMID: 30660504 DOI: 10.1016/j.tcb.2018.12.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/11/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022]
Abstract
Nuclear-import receptors (NIRs) bind nuclear-localization signals (NLSs) of protein cargo in the cytoplasm and transport them into the nucleus. Here, we review advances establishing that NIRs also function in the cytoplasm to prevent and reverse functional and aberrant phase transitions of their cargo, including neurodegenerative disease-linked RNA-binding proteins (RBPs) with prion-like domains, such as TDP-43, FUS, hnRNPA1, and hnRNPA2. NIRs selectively extract cargo from condensed liquid phases thereby regulating functional phase separation. Consequently, NIRs sculpt cytoplasmic membraneless organelles and regulate cellular organization beyond their canonical role in nuclear import. Elevating NIR expression dissolves cytoplasmic RBP aggregates, restores functional RBPs to the nucleus, and rescues disease-linked RBP toxicity. Thus, NIRs could be leveraged therapeutically to restore RBP homeostasis and mitigate neurodegeneration.
Collapse
Affiliation(s)
- Lin Guo
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Joint first authors
| | - Charlotte M Fare
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Joint first authors
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
60
|
RNA Granules and Their Role in Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1203:195-245. [DOI: 10.1007/978-3-030-31434-7_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
61
|
Mutant FUS causes DNA ligation defects to inhibit oxidative damage repair in Amyotrophic Lateral Sclerosis. Nat Commun 2018; 9:3683. [PMID: 30206235 PMCID: PMC6134028 DOI: 10.1038/s41467-018-06111-6] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 08/14/2018] [Indexed: 01/17/2023] Open
Abstract
Genome damage and defective repair are etiologically linked to neurodegeneration. However, the specific mechanisms involved remain enigmatic. Here, we identify defects in DNA nick ligation and oxidative damage repair in a subset of amyotrophic lateral sclerosis (ALS) patients. These defects are caused by mutations in the RNA/DNA-binding protein FUS. In healthy neurons, FUS protects the genome by facilitating PARP1-dependent recruitment of XRCC1/DNA Ligase IIIα (LigIII) to oxidized genome sites and activating LigIII via direct interaction. We discover that loss of nuclear FUS caused DNA nick ligation defects in motor neurons due to reduced recruitment of XRCC1/LigIII to DNA strand breaks. Moreover, DNA ligation defects in ALS patient-derived iPSC lines carrying FUS mutations and in motor neurons generated therefrom are rescued by CRISPR/Cas9-mediated correction of mutation. Our findings uncovered a pathway of defective DNA ligation in FUS-linked ALS and suggest that LigIII-targeted therapies may prevent or slow down disease progression.
Collapse
|
62
|
Ma L, Shi Y, Chen Z, Li S, Zhang J. A novel SETX gene mutation associated with Juvenile amyotrophic lateral sclerosis. Brain Behav 2018; 8:e01066. [PMID: 30052327 PMCID: PMC6160657 DOI: 10.1002/brb3.1066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/24/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023] Open
Abstract
OBJECTS This study aimed to report a novel point mutation associated with juvenile amyotrophic lateral sclerosis (JALS) in a Chinese Han family. METHODS Detailed clinical assessment was applied to two patients, including proband (II-2) and his mother (I-2). Next-generation sequencing (NGS), also known as high-throughput sequencing in whole exon sequence, was performed in the proband to reach the target region. Sanger sequencing was also used to detect DNA sequence variants of the proband and other three members of his family. RESULTS The proband (II-2) and his mother (I-2) were successfully diagnosed according to the clinical manifestations and physical examination. A novel point mutation c.1157T > C in the exon 10 of the SETX gene was identified in II-2 and I-2, resulting in a substitution of methionine (ATG) to threonine (ACG). However, we ultimately did not find the same variant in the other two normal members of his family in addition to 100 unrelated normal subjects. CONCLUSION We presented a novel probably pathogenic missense mutation in exon 10 of SETX gene in a Chinese Han family with JALS.
Collapse
Affiliation(s)
- Limin Ma
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingying Shi
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhongcan Chen
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Shujian Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
63
|
Hikiami R, Yamakado H, Tatsumi S, Ayaki T, Hashi Y, Yamashita H, Sawamoto N, Tsuji T, Urushitani M, Takahashi R. Amyotrophic Lateral Sclerosis after Receiving the Human Papilloma Virus Vaccine: A Case Report of a 15-year-old Girl. Intern Med 2018; 57:1917-1919. [PMID: 29434138 PMCID: PMC6064690 DOI: 10.2169/internalmedicine.0285-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We herein report a 15-year-old girl who developed rapid progressive muscle weakness soon after the third injection of a bivalent human papilloma virus (HPV) vaccine. Although immunotherapies were performed for possible vaccine-related disorders, she died of respiratory failure 14 months after the onset of the disease. A genetic analysis identified a heterozygous p.P525L mutation of the fused in sarcoma (FUS) gene, and a histopathological analysis was also consistent with FUS-associated amyotrophic lateral sclerosis (ALS) without any evidence of neuroinflammation. We concluded the diagnosis to be FUS-ALS, although we cannot completely rule out the possibility that the vaccination worked as a trigger.
Collapse
Affiliation(s)
- Ryota Hikiami
- Department of Neurology, Kyoto University Graduate School of Medicine, Japan
| | - Hodaka Yamakado
- Department of Neurology, Kyoto University Graduate School of Medicine, Japan
| | - Shinsui Tatsumi
- Department of Neurology, Kyoto University Graduate School of Medicine, Japan
| | - Takashi Ayaki
- Department of Neurology, Kyoto University Graduate School of Medicine, Japan
| | - Yuichiro Hashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Japan
| | - Hirofumi Yamashita
- Department of Neurology, Kyoto University Graduate School of Medicine, Japan
| | - Nobukatsu Sawamoto
- Department of Neurology, Kyoto University Graduate School of Medicine, Japan
| | | | - Makoto Urushitani
- Department of Neurology, Kyoto University Graduate School of Medicine, Japan
- Department of Neurology, Shiga University of Medical Science, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Japan
| |
Collapse
|
64
|
Chen H, Kankel MW, Su SC, Han SWS, Ofengeim D. Exploring the genetics and non-cell autonomous mechanisms underlying ALS/FTLD. Cell Death Differ 2018; 25:648-662. [PMID: 29459769 DOI: 10.1038/s41418-018-0060-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022] Open
Abstract
Although amyotrophic lateral sclerosis (ALS), also known as Lou Gehrig's disease, was first described in 1874, a flurry of genetic discoveries in the last 10 years has markedly increased our understanding of this disease. These findings have not only enhanced our knowledge of mechanisms leading to ALS, but also have revealed that ALS shares many genetic causes with another neurodegenerative disease, frontotemporal lobar dementia (FTLD). In this review, we survey how recent genetic studies have bridged our mechanistic understanding of these two related diseases and how the genetics behind ALS and FTLD point to complex disorders, implicating non-neuronal cell types in disease pathophysiology. The involvement of non-neuronal cell types is consistent with a non-cell autonomous component in these diseases. This is further supported by studies that identified a critical role of immune-associated genes within ALS/FTLD and other neurodegenerative disorders. The molecular functions of these genes support an emerging concept that various non-autonomous functions are involved in neurodegeneration. Further insights into such a mechanism(s) will ultimately lead to a better understanding of potential routes of therapeutic intervention. Facts ALS and FTLD are severe neurodegenerative disorders on the same disease spectrum. Multiple cellular processes including dysregulation of RNA homeostasis, imbalance of proteostasis, contribute to ALS/FTLD pathogenesis. Aberrant function in non-neuronal cell types, including microglia, contributes to ALS/FTLD. Strong neuroimmune and neuroinflammatory components are associated with ALS/FTLD patients. Open Questions Why can patients with similar mutations have different disease manifestations, i.e., why do C9ORF72 mutations lead to motor neuron loss in some patients while others exhibit loss of neurons in the frontotemporal lobe? Do ALS causal mutations result in microglial dysfunction and contribute to ALS/FTLD pathology? How do microglia normally act to mitigate neurodegeneration in ALS/FTLD? To what extent do cellular signaling pathways mediate non-cell autonomous communications between distinct central nervous system (CNS) cell types during disease? Is it possible to therapeutically target specific cell types in the CNS?
Collapse
Affiliation(s)
- Hongbo Chen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA, 02115, USA
| | - Mark W Kankel
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Susan C Su
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Steve W S Han
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,GSK, Upper Providence, PA, 19426, USA
| | - Dimitry Ofengeim
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA. .,Sanofi Neuroscience, Framingham, MA, USA.
| |
Collapse
|
65
|
A Regulatory Circuitry Between Gria2, miR-409, and miR-495 Is Affected by ALS FUS Mutation in ESC-Derived Motor Neurons. Mol Neurobiol 2018; 55:7635-7651. [PMID: 29430619 PMCID: PMC6132778 DOI: 10.1007/s12035-018-0884-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/08/2018] [Indexed: 12/12/2022]
Abstract
Mutations in fused in sarcoma (FUS) cause amyotrophic lateral sclerosis (ALS). FUS is a multifunctional protein involved in the biogenesis and activity of several types of RNAs, and its role in the pathogenesis of ALS may involve both direct effects of disease-associated mutations through gain- and loss-of-function mechanisms and indirect effects due to the cross talk between different classes of FUS-dependent RNAs. To explore how FUS mutations impinge on motor neuron-specific RNA-based circuitries, we performed transcriptome profiling of small and long RNAs of motor neurons (MNs) derived from mouse embryonic stem cells carrying a FUS-P517L knock-in mutation, which is equivalent to human FUS-P525L, associated with a severe and juvenile-onset form of ALS. Combining ontological, predictive and molecular analyses, we found an inverse correlation between several classes of deregulated miRNAs and their corresponding mRNA targets in both homozygous and heterozygous P517L MNs. We validated a circuitry in which the upregulation of miR-409-3p and miR-495-3p, belonging to a brain-specific miRNA subcluster implicated in several neurodevelopmental disorders, produced the downregulation of Gria2, a subunit of the glutamate α‐amino‐3‐hydroxy‐5‐methyl-4-isoxazole propionic acid (AMPA) receptor with a significant role in excitatory neurotransmission. Moreover, we found that FUS was involved in mediating such miRNA repression. Gria2 alteration has been proposed to be implicated in MN degeneration, through disturbance of Ca2+ homeostasis, which triggers a cascade of damaging “excitotoxic” events. The molecular cross talk identified highlights a role for FUS in excitotoxicity and in miRNA-dependent regulation of Gria2. This circuitry also proved to be deregulated in heterozygosity, which matches the human condition perfectly.
Collapse
|
66
|
Marrone L, Poser I, Casci I, Japtok J, Reinhardt P, Janosch A, Andree C, Lee HO, Moebius C, Koerner E, Reinhardt L, Cicardi ME, Hackmann K, Klink B, Poletti A, Alberti S, Bickle M, Hermann A, Pandey UB, Hyman AA, Sterneckert JL. Isogenic FUS-eGFP iPSC Reporter Lines Enable Quantification of FUS Stress Granule Pathology that Is Rescued by Drugs Inducing Autophagy. Stem Cell Reports 2018; 10:375-389. [PMID: 29358088 PMCID: PMC5857889 DOI: 10.1016/j.stemcr.2017.12.018] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 12/20/2017] [Accepted: 12/20/2017] [Indexed: 12/12/2022] Open
Abstract
Perturbations in stress granule (SG) dynamics may be at the core of amyotrophic lateral sclerosis (ALS). Since SGs are membraneless compartments, modeling their dynamics in human motor neurons has been challenging, thus hindering the identification of effective therapeutics. Here, we report the generation of isogenic induced pluripotent stem cells carrying wild-type and P525L FUS-eGFP. We demonstrate that FUS-eGFP is recruited into SGs and that P525L profoundly alters their dynamics. With a screening campaign, we demonstrate that PI3K/AKT/mTOR pathway inhibition increases autophagy and ameliorates SG phenotypes linked to P525L FUS by reducing FUS-eGFP recruitment into SGs. Using a Drosophila model of FUS-ALS, we corroborate that induction of autophagy significantly increases survival. Finally, by screening clinically approved drugs for their ability to ameliorate FUS SG phenotypes, we identify a number of brain-penetrant anti-depressants and anti-psychotics that also induce autophagy. These drugs could be repurposed as potential ALS treatments. Generation of isogenic WT and P525L FUS-eGFP reporter iPSCs P525L FUS-eGFP SGs are more numerous, more intense, and larger than WT Increasing PI3K/AKT/mTOR-regulated autophagy reduces FUS-eGFP recruitment to SGs Brain-penetrant drugs that induce autophagy ameliorate the FUS SG phenotype
Collapse
Affiliation(s)
- Lara Marrone
- DFG-Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Ina Poser
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Ian Casci
- Department of Pediatrics, Division of Child Neurology, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Julia Japtok
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Peter Reinhardt
- DFG-Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany; Currently at AbbVie Deutschland GmbH & Co KG, Neuroscience Discovery - Biology Department, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Antje Janosch
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Cordula Andree
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Hyun O Lee
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Claudia Moebius
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Ellen Koerner
- DFG-Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Lydia Reinhardt
- DFG-Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Maria Elena Cicardi
- Department of Pharmacological and Biomolecular Sciences, Centre of Excellence on Neurodegenerative Diseases University of Milan, Milan 20133, Italy
| | - Karl Hackmann
- Currently at AbbVie Deutschland GmbH & Co KG, Neuroscience Discovery - Biology Department, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Barbara Klink
- Institut für Klinische Genetik, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Angelo Poletti
- Department of Pharmacological and Biomolecular Sciences, Centre of Excellence on Neurodegenerative Diseases University of Milan, Milan 20133, Italy
| | - Simon Alberti
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Marc Bickle
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Andreas Hermann
- DFG-Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany; Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE), 01307 Dresden, Germany
| | - Udai Bhan Pandey
- Department of Pediatrics, Division of Child Neurology, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Jared L Sterneckert
- DFG-Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany.
| |
Collapse
|
67
|
Ticozzi N, Silani V. Genotypic and Phenotypic Heterogeneity in Amyotrophic Lateral Sclerosis. NEURODEGENER DIS 2018. [DOI: 10.1007/978-3-319-72938-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
68
|
Lo Bello M, Di Fini F, Notaro A, Spataro R, Conforti FL, La Bella V. ALS-Related Mutant FUS Protein Is Mislocalized to Cytoplasm and Is Recruited into Stress Granules of Fibroblasts from Asymptomatic FUS P525L Mutation Carriers. NEURODEGENER DIS 2017; 17:292-303. [PMID: 29035885 DOI: 10.1159/000480085] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/08/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) shows a strong genetic basis, with SOD1, FUS, TARDBP, and C9ORF72 being the genes most frequently involved. This has allowed identification of asymptomatic mutation carriers, which may be of help in understanding the molecular changes preceding disease onset. OBJECTIVES We studied the cellular expression of FUS protein and the effect of heat-shock- and dithiothreitol-induced stress in fibroblasts from FUS P525L mutation carriers, healthy controls, and patients with sporadic ALS. METHODS Western blots and immunocytochemistry were performed to study the subcellular localization of FUS protein. Control and stressed cells were double stained with FUS and the stress marker TIA-R. RESULTS Fibroblasts from healthy controls and sporadic ALS cases showed a prominent nuclear FUS expression. In the 2 FUS P525L mutation carriers, instead, most cells showed a protein localization in both nucleus and cytoplasm, or exclusively in the cytoplasm. Stress prompted the formation of cytoplasmic granules in all subjects and in sporadic ALS FUS mislocalization to the cytoplasm. Cytoplasmic FUS was recruited into stress granules, which showed a time-dependent decrease in all subjects. However, in the FUS P525L fibroblasts, the granules persisted longer, and they were more numerous than those detected in the cells from controls and sporadic ALS patients. CONCLUSIONS We show that in fibroblasts of FUS P525L mutation carriers, FUS mislocalized to the cytoplasm where it redistributed into stress granules with likely a dose effect, i.e. a higher number of cells with granules, which persist longer, than in controls and ALS cases. These data represent an early molecular change occurring before ALS onset, suggesting a transient preaggregative state.
Collapse
Affiliation(s)
- Margherita Lo Bello
- ALS Clinical Research Center and Laboratory of Neurochemistry, Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | | | | | | | | | | |
Collapse
|
69
|
Kuang L, Kamelgarn M, Arenas A, Gal J, Taylor D, Gong W, Brown M, St Clair D, Kasarskis EJ, Zhu H. Clinical and experimental studies of a novel P525R FUS mutation in amyotrophic lateral sclerosis. NEUROLOGY-GENETICS 2017; 3:e172. [PMID: 28812062 PMCID: PMC5546284 DOI: 10.1212/nxg.0000000000000172] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/16/2017] [Indexed: 12/13/2022]
Abstract
Objective: To describe the clinical features of a novel fused in sarcoma (FUS) mutation in a young adult female amyotrophic lateral sclerosis (ALS) patient with rapid progression of weakness and to experimentally validate the consequences of the P525R mutation in cellular neuronal models. Methods: We conducted sequencing of genomic DNA from the index patient and her family members. Immunocytochemistry was performed in various cellular models to determine whether the newly identified P525R mutant FUS protein accumulated in cytoplasmic inclusions. Clinical features of the index patient were compared with 19 other patients with ALS carrying the P525L mutation in the same amino acid position. Results: A novel mutation c.1574C>G (p.525P>R) in the FUS gene was identified in the index patient. The clinical symptoms are similar to those in familial ALS patients with the P525L mutation at the same position. The P525R mutant FUS protein showed cytoplasmic localization and formed large stress granule–like cytoplasmic inclusions in multiple cellular models. Conclusions: The clinical features of the patient and the cytoplasmic inclusions of the P525R mutant FUS protein strengthen the notion that mutations at position 525 of the FUS protein result in a coherent phenotype characterized by juvenile or young adult onset, rapid progression, variable positive family history, and female preponderance.
Collapse
Affiliation(s)
- Lisha Kuang
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| | - Marisa Kamelgarn
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| | - Alexandra Arenas
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| | - Jozsef Gal
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| | - Deborah Taylor
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| | - Weiming Gong
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| | - Martin Brown
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| | - Daret St Clair
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| | - Edward J Kasarskis
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| | - Haining Zhu
- Molecular and Cellular Biochemistry (L.K., J.G., H.Z.), Department of Toxicology and Cancer Biology (M.K., A.A., D.S.C., H.Z.), and Department of Neurology (D.T., E.J.K.), College of Medicine, University of Kentucky, Lexington; Hefei National Laboratory for Physical Sciences at the Microscale (W.G.), University of Science and Technology of China, Anhui; Department of Neurology (M.B.), University of Louisville; and Research and Development (E.J.K., H.Z.), Lexington VA Medical Center, KY
| |
Collapse
|
70
|
Affiliation(s)
- Robert H Brown
- From the Department of Neurology, University of Massachusetts Medical School, Worcester (R.H.B.); and the Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, King's College London, London (A.A.-C.)
| | - Ammar Al-Chalabi
- From the Department of Neurology, University of Massachusetts Medical School, Worcester (R.H.B.); and the Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, King's College London, London (A.A.-C.)
| |
Collapse
|
71
|
Scekic-Zahirovic J, Oussini HE, Mersmann S, Drenner K, Wagner M, Sun Y, Allmeroth K, Dieterlé S, Sinniger J, Dirrig-Grosch S, René F, Dormann D, Haass C, Ludolph AC, Lagier-Tourenne C, Storkebaum E, Dupuis L. Motor neuron intrinsic and extrinsic mechanisms contribute to the pathogenesis of FUS-associated amyotrophic lateral sclerosis. Acta Neuropathol 2017; 133:887-906. [PMID: 28243725 PMCID: PMC5427169 DOI: 10.1007/s00401-017-1687-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/08/2017] [Accepted: 02/16/2017] [Indexed: 12/11/2022]
Abstract
Motor neuron-extrinsic mechanisms have been shown to participate in the pathogenesis of ALS-SOD1, one familial form of amyotrophic lateral sclerosis (ALS). It remains unclear whether such mechanisms contribute to other familial forms, such as TDP-43 and FUS-associated ALS. Here, we characterize a single-copy mouse model of ALS-FUS that conditionally expresses a disease-relevant truncating FUS mutant from the endogenous murine Fus gene. We show that these mice, but not mice heterozygous for a Fus null allele, develop similar pathology as ALS-FUS patients and a mild motor neuron phenotype. Most importantly, CRE-mediated rescue of the Fus mutation within motor neurons prevented degeneration of motor neuron cell bodies, but only delayed appearance of motor symptoms. Indeed, we observed downregulation of multiple myelin-related genes, and increased numbers of oligodendrocytes in the spinal cord supporting their contribution to behavioral deficits. In all, we show that mutant FUS triggers toxic events in both motor neurons and neighboring cells to elicit motor neuron disease.
Collapse
|
72
|
Abstract
Paediatric motor neuron diseases encompass a group of neurodegenerative diseases characterised by the onset of muscle weakness and atrophy before the age of 18 years, attributable to motor neuron loss across various neuronal networks in the brain and spinal cord. While the genetic underpinnings are diverse, advances in next generation sequencing have transformed diagnostic paradigms. This has reinforced the clinical phenotyping and molecular genetic expertise required to navigate the complexities of such diagnoses. In turn, improved genetic technology and subsequent gene identification have enabled further insights into the mechanisms of motor neuron degeneration and how these diseases form part of a neurodegenerative disorder spectrum. Common pathophysiologies include abnormalities in axonal architecture and function, RNA processing, and protein quality control. This review incorporates an overview of the clinical manifestations, genetics, and pathophysiology of inherited paediatric motor neuron disorders beyond classic SMN1-related spinal muscular atrophy and describes recent advances in next generation sequencing and its clinical application. Specific disease-modifying treatment is becoming a clinical reality in some disorders of the motor neuron highlighting the importance of a timely and specific diagnosis.
Collapse
|
73
|
Corcia P, Couratier P, Blasco H, Andres C, Beltran S, Meininger V, Vourc’h P. Genetics of amyotrophic lateral sclerosis. Rev Neurol (Paris) 2017; 173:254-262. [DOI: 10.1016/j.neurol.2017.03.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 03/27/2017] [Indexed: 12/12/2022]
|
74
|
Liu ZJ, Lin HX, Liu GL, Tao QQ, Ni W, Xiao BG, Wu ZY. The investigation of genetic and clinical features in Chinese patients with juvenile amyotrophic lateral sclerosis. Clin Genet 2017; 92:267-273. [PMID: 28429524 DOI: 10.1111/cge.13015] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/08/2017] [Accepted: 03/10/2017] [Indexed: 12/13/2022]
Abstract
Juvenile amyotrophic lateral sclerosis (JALS) occurs at an age of onset below 25 years with a heterogeneous disease onset location, variable progression and survival time. To investigate whether an ALS gene profile could resolve any aspects of clinical symptom heterogeneity, we have used targeted sequencing technology in a cohort of 12 JALS patients of Chinese descent. We detected 5 likely pathogenic mutations, 2 in familial probands and 3 in sporadic patients. One was a known TARDBP mutation (p.G348V) and 4 were FUS frameshift mutations including a known p.Gln519Ilefs*9 mutation and 3 novel mutations, p.Gly515Valfs*14, p.Gly486Profs*30, and p.Arg498Alafs*32. Of the 4 FUS mutations, 2 were able to be confirmed as de novo mutations. The TARDBP mutation carrier showed a classic ALS phenotype. All patients with FUS mutations experienced limb weakness at an early age and developed bulbar symptoms during the disease course. FUS mutations have previously been associated with increased JALS disease progression, however, we found a large range 12 to 84 months in disease survival (mean 58.2 months). Our results justify future screening for variants in FUS as it remains the most frequent genetic determinant of early onset, JALS (found in 30% of our patients).
Collapse
Affiliation(s)
- Z-J Liu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - H-X Lin
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and The Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China.,Department of Neurology and Institute of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - G-L Liu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and The Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China
| | - Q-Q Tao
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and The Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China
| | - W Ni
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and The Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, China
| | - B-G Xiao
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Z-Y Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
75
|
Screening of SOD1, FUS and TARDBP genes in patients with amyotrophic lateral sclerosis in central-southern China. Sci Rep 2016; 6:32478. [PMID: 27604643 PMCID: PMC5015023 DOI: 10.1038/srep32478] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/08/2016] [Indexed: 01/26/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting motor neurons of the brain, brainstem and spinal cord. To date, mutations in more than 30 genes have been linked to the pathogenesis of ALS. Among them, SOD1, FUS and TARDBP are ranked as the three most common genes associated with ALS. However, no mutation analysis has been reported in central-southern China. In this study, we sequenced SOD1, FUS and TARDBP in a central-southern Chinese cohort of 173 patients with ALS (15 familial ALS and 158 sporadic ALS) to detect mutations. As a result, five missense mutations in SOD1, namely, p.D101N, p.D101G, p.C111Y, p.N86S and p.V87A, were identified in three unrelated familial probands and three sporadic cases; two mutations in FUS were found in two unrelated familial probands, including an insertion mutation (p.P525_Y526insY) and a missense mutation (p.R521H); no variants of TARDBP were observed in patients. Therefore, SOD1 mutations were present in 20.0% of familial ALS patients and 1.9% of sporadic ALS patients, while FUS mutations were responsible for 13.3% of familial ALS cases, and TARDBP mutations were rare in either familial or sporadic ALS cases. This study broadens the known mutational spectrum in patients with ALS and further demonstrates the necessity for genetic screening in ALS patients from central-southern China.
Collapse
|
76
|
Zou ZY, Liu MS, Li XG, Cui LY. Mutations in FUS are the most frequent genetic cause in juvenile sporadic ALS patients of Chinese origin. Amyotroph Lateral Scler Frontotemporal Degener 2016; 17:249-52. [PMID: 26972116 DOI: 10.3109/21678421.2016.1143012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Juvenile onset ALS is a very rare form of motor neuron disease, with the first symptoms of motor neuron degeneration manifested before 25 years of age. Mutations in the alsin (ALS2), senataxin (SETX), and spatacsin (SPG11) genes have been associated with familial ALS with juvenile onset and slow progression, whereas the genetic architecture of sporadic juvenile ALS remains unclear. We screened mutations in C9orf72, SOD1, FUS, TARDBP, ANG, VCP and PFN1 in 16 juvenile sporadic ALS patients. Four cases (25%) carrying FUS mutations and one individual (6%) harbouring a SOD1 mutation were identified. All cases had an aggressive disease course. Our results suggest that FUS mutations are the most frequent genetic cause in early-onset sporadic ALS patients of Chinese origin. Genetic testing of FUS should be performed in early-onset ALS patients especially those with an aggressive disease course.
Collapse
Affiliation(s)
- Zhang-Yu Zou
- a Department of Neurology , Fujian Medical University Union Hospital , Fuzhou and.,b Department of Neurology , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Ming-Sheng Liu
- b Department of Neurology , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Xiao-Guang Li
- b Department of Neurology , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Li-Ying Cui
- b Department of Neurology , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| |
Collapse
|
77
|
Leblond CS, Webber A, Gan-Or Z, Moore F, Dagher A, Dion PA, Rouleau GA. De novo FUS P525L mutation in Juvenile amyotrophic lateral sclerosis with dysphonia and diplopia. NEUROLOGY-GENETICS 2016; 2:e63. [PMID: 27123482 PMCID: PMC4830191 DOI: 10.1212/nxg.0000000000000063] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 01/27/2016] [Indexed: 12/12/2022]
Abstract
Juvenile amyotrophic lateral sclerosis (jALS) is characterized by progressive upper and lower motor neuron degeneration leading to facial muscle spasticity, spastic dysarthria, and spastic gait with an early onset (before 25 years old). Unlike adult-onset amyotrophic lateral sclerosis (ALS), patients with jALS tend to have slower progression of motor neuron disease and prolonged survival to a normal life expectancy. Mutations in FUS gene have been reported in jALS,(1) including p.P525L mutation that has been consistently associated with early onset and aggressive presentation.(2) Here, we report a patient carrying p.P525L FUS mutation and experiencing an aggressive course of ALS presenting with dysphonia and diplopia.
Collapse
Affiliation(s)
- Claire S Leblond
- Department of Human Genetics (C.S.L., Z.G.-O.), Montreal Neurological Institute and Hospital (C.S.L., A.W., Z.G.-O., F.M., A.D., P.A.D., G.A.R.), and Department of Neurology and Neurosurgery (A.W., F.M., A.D., P.A.D., G.A.R.), McGill University, Montreal, Quebec, Canada
| | - Alina Webber
- Department of Human Genetics (C.S.L., Z.G.-O.), Montreal Neurological Institute and Hospital (C.S.L., A.W., Z.G.-O., F.M., A.D., P.A.D., G.A.R.), and Department of Neurology and Neurosurgery (A.W., F.M., A.D., P.A.D., G.A.R.), McGill University, Montreal, Quebec, Canada
| | - Ziv Gan-Or
- Department of Human Genetics (C.S.L., Z.G.-O.), Montreal Neurological Institute and Hospital (C.S.L., A.W., Z.G.-O., F.M., A.D., P.A.D., G.A.R.), and Department of Neurology and Neurosurgery (A.W., F.M., A.D., P.A.D., G.A.R.), McGill University, Montreal, Quebec, Canada
| | - Fraser Moore
- Department of Human Genetics (C.S.L., Z.G.-O.), Montreal Neurological Institute and Hospital (C.S.L., A.W., Z.G.-O., F.M., A.D., P.A.D., G.A.R.), and Department of Neurology and Neurosurgery (A.W., F.M., A.D., P.A.D., G.A.R.), McGill University, Montreal, Quebec, Canada
| | - Alain Dagher
- Department of Human Genetics (C.S.L., Z.G.-O.), Montreal Neurological Institute and Hospital (C.S.L., A.W., Z.G.-O., F.M., A.D., P.A.D., G.A.R.), and Department of Neurology and Neurosurgery (A.W., F.M., A.D., P.A.D., G.A.R.), McGill University, Montreal, Quebec, Canada
| | - Patrick A Dion
- Department of Human Genetics (C.S.L., Z.G.-O.), Montreal Neurological Institute and Hospital (C.S.L., A.W., Z.G.-O., F.M., A.D., P.A.D., G.A.R.), and Department of Neurology and Neurosurgery (A.W., F.M., A.D., P.A.D., G.A.R.), McGill University, Montreal, Quebec, Canada
| | - Guy A Rouleau
- Department of Human Genetics (C.S.L., Z.G.-O.), Montreal Neurological Institute and Hospital (C.S.L., A.W., Z.G.-O., F.M., A.D., P.A.D., G.A.R.), and Department of Neurology and Neurosurgery (A.W., F.M., A.D., P.A.D., G.A.R.), McGill University, Montreal, Quebec, Canada
| |
Collapse
|
78
|
Sharma A, Lyashchenko AK, Lu L, Nasrabady SE, Elmaleh M, Mendelsohn M, Nemes A, Tapia JC, Mentis GZ, Shneider NA. ALS-associated mutant FUS induces selective motor neuron degeneration through toxic gain of function. Nat Commun 2016; 7:10465. [PMID: 26842965 PMCID: PMC4742863 DOI: 10.1038/ncomms10465] [Citation(s) in RCA: 252] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/14/2015] [Indexed: 12/13/2022] Open
Abstract
Mutations in FUS cause amyotrophic lateral sclerosis (ALS), including some of the most aggressive, juvenile-onset forms of the disease. FUS loss-of-function and toxic gain-of-function mechanisms have been proposed to explain how mutant FUS leads to motor neuron degeneration, but neither has been firmly established in the pathogenesis of ALS. Here we characterize a series of transgenic FUS mouse lines that manifest progressive, mutant-dependent motor neuron degeneration preceded by early, structural and functional abnormalities at the neuromuscular junction. A novel, conditional FUS knockout mutant reveals that postnatal elimination of FUS has no effect on motor neuron survival or function. Moreover, endogenous FUS does not contribute to the onset of the ALS phenotype induced by mutant FUS. These findings demonstrate that FUS-dependent motor degeneration is not due to loss of FUS function, but to the gain of toxic properties conferred by ALS mutations. The mechanism by which FUS mutations cause familial ALS remains unclear. Here, the authors use mouse transgenic models to show that a toxic gain-of-function underlies motor neuron degeneration, and that the toxicity of mutant FUS does not depend on a loss or excess of FUS activity.
Collapse
Affiliation(s)
- Aarti Sharma
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, 630 W 168th Street, P&S Building, Room 5-423, New York, New York 10032, USA
| | - Alexander K Lyashchenko
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, 630 W 168th Street, P&S Building, Room 5-423, New York, New York 10032, USA
| | - Lei Lu
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, 630 W 168th Street, P&S Building, Room 5-423, New York, New York 10032, USA
| | - Sara Ebrahimi Nasrabady
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, New York 10032, USA
| | - Margot Elmaleh
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, 630 W 168th Street, P&S Building, Room 5-423, New York, New York 10032, USA
| | - Monica Mendelsohn
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, New York 10032, USA
| | - Adriana Nemes
- Department of Neuroscience, Howard Hughes Medical Institute, Columbia University, New York, New York 10032, USA
| | - Juan Carlos Tapia
- Department of Neuroscience, Columbia University, New York, New York 10032, USA
| | - George Z Mentis
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, New York 10032, USA
| | - Neil A Shneider
- Department of Neurology, Center for Motor Neuron Biology and Disease, Columbia University, 630 W 168th Street, P&S Building, Room 5-423, New York, New York 10032, USA
| |
Collapse
|
79
|
Hirayanagi K, Sato M, Furuta N, Makioka K, Ikeda Y. Juvenile-onset Sporadic Amyotrophic Lateral Sclerosis with a Frameshift FUS Gene Mutation Presenting Unique Neuroradiological Findings and Cognitive Impairment. Intern Med 2016; 55:689-93. [PMID: 26984092 DOI: 10.2169/internalmedicine.55.5569] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A 24-year-old Japanese woman developed anterocollis, weakness of the proximal arms, and subsequent cognitive impairment. A neurological examination revealed amyotrophic lateral sclerosis (ALS) without a family history. Systemic muscle atrophy progressed rapidly. Cerebral MRI clearly exhibited high signal intensities along the bilateral pyramidal tracts. An analysis of the FUS gene revealed a heterozygous two-base pair deletion, c.1507-1508delAG (p.G504WfsX515). A subset of juvenile-onset familial/sporadic ALS cases with FUS gene mutations reportedly demonstrates mental retardation or learning difficulty. Our study emphasizes the importance of conducting a FUS gene analysis in juvenile-onset ALS cases, even when no family occurrence is confirmed.
Collapse
|
80
|
Nakamura R, Sone J, Atsuta N, Tohnai G, Watanabe H, Yokoi D, Nakatochi M, Watanabe H, Ito M, Senda J, Katsuno M, Tanaka F, Li Y, Izumi Y, Morita M, Taniguchi A, Kano O, Oda M, Kuwabara S, Abe K, Aiba I, Okamoto K, Mizoguchi K, Hasegawa K, Aoki M, Hattori N, Tsuji S, Nakashima K, Kaji R, Sobue G. Next-generation sequencing of 28 ALS-related genes in a Japanese ALS cohort. Neurobiol Aging 2015; 39:219.e1-8. [PMID: 26742954 DOI: 10.1016/j.neurobiolaging.2015.11.030] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/23/2015] [Accepted: 11/28/2015] [Indexed: 10/22/2022]
Abstract
We investigated the frequency and contribution of variants of the 28 known amyotrophic lateral sclerosis (ALS)-related genes in Japanese ALS patients. We designed a multiplex, polymerase chain reaction-based primer panel to amplify the coding regions of the 28 ALS-related genes and sequenced DNA samples from 257 Japanese ALS patients using an Ion Torrent PGM sequencer. We also performed exome sequencing and identified variants of the 28 genes in an additional 251 ALS patients using an Illumina HiSeq 2000 platform. We identified the known ALS pathogenic variants and predicted the functional properties of novel nonsynonymous variants in silico. These variants were confirmed by Sanger sequencing. Known pathogenic variants were identified in 19 (48.7%) of the 39 familial ALS patients and 14 (3.0%) of the 469 sporadic ALS patients. Thirty-two sporadic ALS patients (6.8%) harbored 1 or 2 novel nonsynonymous variants of ALS-related genes that might be deleterious. This study reports the first extensive genetic screening of Japanese ALS patients. These findings are useful for developing genetic screening and counseling strategies for such patients.
Collapse
Affiliation(s)
- Ryoichi Nakamura
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun Sone
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoki Atsuta
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Genki Tohnai
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hazuki Watanabe
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Neurology, Japanese Red Cross Nagoya Daiichi Hospital, Nagoya, Japan
| | - Daichi Yokoi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Nakatochi
- Bioinformatics Section, Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| | - Mizuki Ito
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jo Senda
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Neurology, Komaki City Hospital, Komaki, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuanzhe Li
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yuishin Izumi
- Department of Neurology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Akira Taniguchi
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Osamu Kano
- Division of Neurology, Department of Internal Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Masaya Oda
- Department of Neurology, Vihara Hananosato Hospital, Miyoshi, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koji Abe
- Department of Neurology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Ikuko Aiba
- Department of Neurology, National Hospital Organization Higashinagoya National Hospital, Nagoya, Japan
| | - Koichi Okamoto
- Department of Neurology, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Kouichi Mizoguchi
- Department of Neurology, National Hospital Organization Shizuoka-Fuji Hospital, Fujinomiya, Japan
| | - Kazuko Hasegawa
- Division of Neurology, National Hospital Organization, Sagamihara National Hospital, Sagamihara, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Shoji Tsuji
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenji Nakashima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ryuji Kaji
- Department of Neurology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | | |
Collapse
|
81
|
Treatment with a Global Methyltransferase Inhibitor Induces the Intranuclear Aggregation of ALS-Linked FUS Mutant In Vitro. Neurochem Res 2015; 41:826-35. [PMID: 26603295 DOI: 10.1007/s11064-015-1758-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/01/2015] [Accepted: 11/03/2015] [Indexed: 10/22/2022]
Abstract
FUS/TLS (fused in sarcoma/translocated in liposarcoma) encodes a multifunctional DNA/RNA binding protein with non-classical carboxy (C)-terminal nuclear localization signal (NLS). A variety of ALS-linked mutations are clustered in the C-terminal NLS, resulting in the cytoplasmic mislocalization and aggregation. Since the arginine methylations are implicated in the nuclear-cytoplasmic shuttling of FUS, a methylation inhibitor could be one of therapeutic targets for FUS-linked ALS. We here examined effects of methylation inhibitors on the cytoplasmic mislocalization and aggregates of ALS-linked C-terminal FUS mutant in a cell culture system. Treatment with adenosine dialdehyde (AdOx), a representative global methyltransferase inhibitor, remarkably mitigated the cytoplasmic mislocalization and aggregation of FUS mutant, which is consistent with previous reports. However, AdOx treatment of higher concentration and longer time period evoked the intranuclear aggregation of the ectopic expressed FUS protein. The pull down assay and the morphological analysis indicated the binding between FUS and Transportin could be potentiated by AdOx treatment through modulating methylation status in RGG domains of FUS. These findings indicated the treatment with a methylation inhibitor at the appropriate levels could alleviate the cytoplasmic mislocalization but in excess this could cause the intranuclear aggregation of FUS C-terminal mutant.
Collapse
|
82
|
Theme 9 Genetics. Amyotroph Lateral Scler Frontotemporal Degener 2015; 16 Suppl 1:160-75. [DOI: 10.3109/21678421.2015.1098814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
83
|
Zou ZY, Liu MS, Li XG, Cui LY. Mutations in SOD1 and FUS caused juvenile-onset sporadic amyotrophic lateral sclerosis with aggressive progression. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:221. [PMID: 26488017 DOI: 10.3978/j.issn.2305-5839.2015.09.04] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Juvenile onset amyotrophic lateral sclerosis (ALS) is a very rare form of motor neuron disease, with the first symptoms of motor neuron degeneration manifested before 25 years of age. Juvenile ALS is more frequently familial in nature than the adult-onset forms. Mutations in the alsin (ALS2), senataxin (SETX), and Spatacsin (SPG11) have been associated with familial ALS with juvenile onset and slowly progression. Here we reported two apparently sporadic ALS with juvenile onset and aggressive progression caused by mutations in the SOD1 and FUS gene. We also reviewed juvenile-onset ALS in publications. Our findings, together with other researches, confirms that both SOD1 and FUS mutations can lead to juvenile-onset malignant form of ALS and should be screened in ALS patients with an earlier age of onset, aggressive progression, even if there is no apparent family history.
Collapse
Affiliation(s)
- Zhang-Yu Zou
- 1 Department of Neurology, Fujian Medical University Union Hospital, Fuzhou 350001, China ; 2 Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Ming-Sheng Liu
- 1 Department of Neurology, Fujian Medical University Union Hospital, Fuzhou 350001, China ; 2 Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Xiao-Guang Li
- 1 Department of Neurology, Fujian Medical University Union Hospital, Fuzhou 350001, China ; 2 Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Li-Ying Cui
- 1 Department of Neurology, Fujian Medical University Union Hospital, Fuzhou 350001, China ; 2 Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
84
|
Darovic S, Prpar Mihevc S, Župunski V, Gunčar G, Štalekar M, Lee YB, Shaw CE, Rogelj B. Phosphorylation of C-terminal tyrosine residue 526 in FUS impairs its nuclear import. J Cell Sci 2015; 128:4151-9. [PMID: 26403203 DOI: 10.1242/jcs.176602] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/17/2015] [Indexed: 12/13/2022] Open
Abstract
Aberrant cytoplasmic aggregation of FUS, which is caused by mutations primarily in the C-terminal nuclear localisation signal, is associated with 3% of cases of familial amyotrophic lateral sclerosis (ALS). FUS aggregates are also pathognomonic for 10% of all frontotemporal lobar degeneration (FTLD) cases; however, these cases are not associated with mutations in the gene encoding FUS. This suggests that there are differences in the mechanisms that drive inclusion formation of FUS in ALS and FTLD. Here, we show that the C-terminal tyrosine residue at position 526 of FUS is crucial for normal nuclear import. This tyrosine is subjected to phosphorylation, which reduces interaction with transportin 1 and might consequentially affect the transport of FUS into the nucleus. Furthermore, we show that this phosphorylation can occur through the activity of the Src family of kinases. Our study implicates phosphorylation as an additional mechanism by which nuclear transport of FUS might be regulated and potentially perturbed in ALS and FTLD.
Collapse
Affiliation(s)
- Simona Darovic
- Jožef Stefan Institute, Department of Biotechnology, Jamova 39, Ljubljana 1000, Slovenia
| | - Sonja Prpar Mihevc
- Jožef Stefan Institute, Department of Biotechnology, Jamova 39, Ljubljana 1000, Slovenia
| | - Vera Župunski
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Gregor Gunčar
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Maja Štalekar
- Jožef Stefan Institute, Department of Biotechnology, Jamova 39, Ljubljana 1000, Slovenia
| | - Youn-Bok Lee
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, King's College London, London SE5 9RT, UK
| | - Christopher E Shaw
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, King's College London, London SE5 9RT, UK
| | - Boris Rogelj
- Jožef Stefan Institute, Department of Biotechnology, Jamova 39, Ljubljana 1000, Slovenia Biomedical Research Institute BRIS, Puhova 10, Ljubljana 1000, Slovenia
| |
Collapse
|
85
|
Hübers A, Just W, Rosenbohm A, Müller K, Marroquin N, Goebel I, Högel J, Thiele H, Altmüller J, Nürnberg P, Weishaupt JH, Kubisch C, Ludolph AC, Volk AE. De novo FUS mutations are the most frequent genetic cause in early-onset German ALS patients. Neurobiol Aging 2015; 36:3117.e1-3117.e6. [PMID: 26362943 DOI: 10.1016/j.neurobiolaging.2015.08.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/15/2015] [Accepted: 08/08/2015] [Indexed: 01/27/2023]
Abstract
In amyotrophic lateral sclerosis (ALS) patients with known genetic cause, mutations in chromosome 9 open reading frame 72 (C9orf72) and superoxide dismutase 1 (SOD1) account for most familial and late-onset sporadic cases, whereas mutations in fused in sarcoma (FUS) can be identified in just around 5% of familial and 1% of overall sporadic cases. There are only few reports on de novo FUS mutations in juvenile ALS patients. To date, no systematic evaluation on the frequency of de novo FUS mutations in early-onset ALS patients has been conducted. Here, we screened a cohort of 14 early-onset sporadic ALS patients (onset age <35 years) to determine the frequency of mutations in C9orf72, SOD1, and FUS in this defined patient cohort. All patients were recruited prospectively by a single center in a period of 38 months. No mutations were detected in SOD1 or C9orf72; however, we identified 6 individuals (43%) carrying a heterozygous FUS mutation including 1 mutation that has not been described earlier (c.1504delG [p.Asp502Thrfs*27]). Genetic testing of parents was possible in 5 families and revealed that the mutations in these patients arose de novo. Three of the 6 identified patients presented with initial bulbar symptoms. Our study identifies FUS mutations as the most frequent genetic cause in early-onset ALS. Genetic testing of FUS thus seems indicated in sporadic early-onset ALS patients especially if showing predominant bulbar symptoms and an aggressive disease course.
Collapse
Affiliation(s)
- Annemarie Hübers
- Department of Neurology, University Hospital of Ulm, Ulm, Germany.
| | - Walter Just
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Angela Rosenbohm
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Kathrin Müller
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | | | - Ingrid Goebel
- Institute of Human Genetics, University of Ulm, Ulm, Germany; Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Josef Högel
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Holger Thiele
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Janine Altmüller
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany; Institute of Human Genetics, University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | | | - Christian Kubisch
- Institute of Human Genetics, University of Ulm, Ulm, Germany; Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Albert C Ludolph
- Department of Neurology, University Hospital of Ulm, Ulm, Germany
| | - Alexander E Volk
- Institute of Human Genetics, University of Ulm, Ulm, Germany; Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
86
|
Coady TH, Manley JL. ALS mutations in TLS/FUS disrupt target gene expression. Genes Dev 2015; 29:1696-706. [PMID: 26251528 PMCID: PMC4561479 DOI: 10.1101/gad.267286.115] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/22/2015] [Indexed: 12/12/2022]
Abstract
In this study, Coadey et al. investigated how mutations in the RNA/DNA-binding protein TLS/FUS (FUS), caused by ALS, affect target gene expression. They used several FUS derivatives with ALS mutations and showed that FUS-containing aggregates can alter gene expression by a toxic gain-of-function mechanism. These findings establish that ALS mutations in FUS can strongly impact target gene expression. Amyotrophic lateral sclerosis (ALS) is caused by mutations in a number of genes, including the gene encoding the RNA/DNA-binding protein translocated in liposarcoma or fused in sarcoma (TLS/FUS or FUS). Previously, we identified a number of FUS target genes, among them MECP2. To investigate how ALS mutations in FUS might impact target gene expression, we examined the effects of several FUS derivatives harboring ALS mutations, such as R521C (FUSC), on MECP2 expression in transfected human U87 cells. Strikingly, FUSC and other mutants not only altered MECP2 alternative splicing but also markedly increased mRNA abundance, which we show resulted from sharply elevated stability. Paradoxically, however, MeCP2 protein levels were significantly reduced in cells expressing ALS mutant derivatives. Providing a parsimonious explanation for these results, biochemical fractionation and in vivo localization studies revealed that MECP2 mRNA colocalized with cytoplasmic FUSC in insoluble aggregates, which are characteristic of ALS mutant proteins. Together, our results establish that ALS mutations in FUS can strongly impact target gene expression, reflecting a dominant effect of FUS-containing aggregates.
Collapse
Affiliation(s)
- Tristan H Coady
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - James L Manley
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| |
Collapse
|
87
|
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common adult-onset motor neuron disease. It is characterized by neuronal loss and degeneration of the upper motor neurons (UMNs) and lower motor neurons (LMNs), and is usually fatal due to respiratory failure within 3–5 years of onset. Although approximately 5–10 % of patients with ALS have an inherited form of the disease, the distinction between hereditary and apparently sporadic ALS (SALS) seems to be artificial. Thus, genetic factors play a role in all types of ALS, to a greater or lesser extent. During the decade of upheaval, the evolution of molecular genetics technology has rapidly advanced our genetic knowledge about the causes of ALS, and the relationship between the genetic subtypes and clinical phenotype. In this review, we will focus on the possible genotype-phenotype correlation in hereditary ALS. Uncovering the identity of the genetic factors in ALS will not only improve the accuracy of ALS diagnosis, but may also provide new approaches for preventing and treating the disease.
Collapse
Affiliation(s)
- Satoshi Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556 Japan
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556 Japan
| |
Collapse
|
88
|
Tarlarini C, Lunetta C, Mosca L, Avemaria F, Riva N, Mantero V, Maestri E, Quattrini A, Corbo M, Melazzini MG, Penco S. Novel FUS mutations identified through molecular screening in a large cohort of familial and sporadic amyotrophic lateral sclerosis. Eur J Neurol 2015; 22:1474-81. [DOI: 10.1111/ene.12772] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/27/2015] [Indexed: 02/02/2023]
Affiliation(s)
- C. Tarlarini
- Medical Genetics Unit; Department of Laboratory Medicine; Niguarda Ca' Granda Hospital; Milan Italy
| | - C. Lunetta
- NEuroMuscular Omnicentre (NEMO); Fondazione Serena Onlus; Niguarda Ca' Granda Hospital; Milan Italy
| | - L. Mosca
- Medical Genetics Unit; Department of Laboratory Medicine; Niguarda Ca' Granda Hospital; Milan Italy
| | - F. Avemaria
- Medical Genetics Unit; Department of Laboratory Medicine; Niguarda Ca' Granda Hospital; Milan Italy
| | - N. Riva
- Neuropathology Unit; Institute of Experimental Neurology and Division of Neuroscience; IRCCS San Raffaele Scientific Institute; Milan Italy
| | - V. Mantero
- Neurological Department; A. Manzoni Hospital; Lecco Italy
| | - E. Maestri
- NEuroMuscular Omnicentre (NEMO); Fondazione Serena Onlus; Niguarda Ca' Granda Hospital; Milan Italy
| | - A. Quattrini
- Neuropathology Unit; Institute of Experimental Neurology and Division of Neuroscience; IRCCS San Raffaele Scientific Institute; Milan Italy
| | - M. Corbo
- NEuroMuscular Omnicentre (NEMO); Fondazione Serena Onlus; Niguarda Ca' Granda Hospital; Milan Italy
- Department of Neurorehabilitation Sciences; Casa Cura Policlinico; Milan Italy
| | - M. G. Melazzini
- NEuroMuscular Omnicentre (NEMO); Fondazione Serena Onlus; Niguarda Ca' Granda Hospital; Milan Italy
| | - S. Penco
- Medical Genetics Unit; Department of Laboratory Medicine; Niguarda Ca' Granda Hospital; Milan Italy
| |
Collapse
|
89
|
Marangi G, Traynor BJ. Genetic causes of amyotrophic lateral sclerosis: new genetic analysis methodologies entailing new opportunities and challenges. Brain Res 2015; 1607:75-93. [PMID: 25316630 PMCID: PMC5916786 DOI: 10.1016/j.brainres.2014.10.009] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/03/2014] [Accepted: 10/05/2014] [Indexed: 12/11/2022]
Abstract
The genetic architecture of amyotrophic lateral sclerosis (ALS) is being increasingly understood. In this far-reaching review, we examine what is currently known about ALS genetics and how these genes were initially identified. We also discuss the various types of mutations that might underlie this fatal neurodegenerative condition and outline some of the strategies that might be useful in untangling them. These include expansions of short repeat sequences, common and low-frequency genetic variations, de novo mutations, epigenetic changes, somatic mutations, epistasis, oligogenic and polygenic hypotheses. This article is part of a Special Issue entitled ALS complex pathogenesis.
Collapse
Affiliation(s)
- Giuseppe Marangi
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA; Institute of Medical Genetics, Catholic University, Roma, Italy.
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
90
|
Liu X, Chen J, liu W, Li X, Chen Q, Liu T, Gao S, Deng M. The fused in sarcoma protein forms cytoplasmic aggregates in motor neurons derived from integration-free induced pluripotent stem cells generated from a patient with familial amyotrophic lateral sclerosis carrying the FUS-P525L mutation. Neurogenetics 2015; 16:223-31. [DOI: 10.1007/s10048-015-0448-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/14/2015] [Indexed: 12/17/2022]
|
91
|
Kent L, Vizard TN, Smith BN, Topp SD, Vance C, Gkazi A, Miller J, Shaw CE, Talbot K. Autosomal dominant inheritance of rapidly progressive amyotrophic lateral sclerosis due to a truncation mutation in the fused in sarcoma (FUS) gene. Amyotroph Lateral Scler Frontotemporal Degener 2014; 15:557-62. [PMID: 24899262 DOI: 10.3109/21678421.2014.920033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 04/28/2014] [Indexed: 12/12/2022]
Abstract
Mutations in the gene encoding the RNA-binding protein fused in sarcoma (FUS) account for 4 - 5% of familial cases of amyotrophic lateral sclerosis (ALS). We describe the identification and in vitro cellular characterization of a genetic mutation in a family in which the index case, and subsequently her two children, each developed rapidly progressive ALS at a young age and died within a year of onset. Exome capture and sequencing revealed a mutation in the FUS gene consisting of a 2-bp deletion, c.1509_1510delAG, resulting in a predicted truncated protein, p.G504Wfs * 12, lacking the nuclear localization signal. Expression of this mutation in HEK293 and NSC-34 cells demonstrated severe cytoplasmic mislocalization of mutant FUS, and colocalization with stress granules when compared to wild-type, R521C and P525L mutant FUS. This study provides further evidence of a broad correlation between clinical severity of FUS-related ALS and mislocalization of the protein to the cytoplasm.
Collapse
Affiliation(s)
- Louisa Kent
- Nuffield Department of Clinical Neurosciences, University of Oxford
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Abstract
The RNA-binding protein fused-in-sarcoma (FUS) has been associated with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), two neurodegenerative disorders that share similar clinical and pathological features. Both missense mutations and overexpression of wild-type FUS protein can be pathogenic in human patients. To study the molecular and cellular basis by which FUS mutations and overexpression cause disease, we generated novel transgenic mice globally expressing low levels of human wild-type protein (FUS(WT)) and a pathological mutation (FUS(R521G)). FUS(WT) and FUS(R521G) mice that develop severe motor deficits also show neuroinflammation, denervated neuromuscular junctions, and premature death, phenocopying the human diseases. A portion of FUS(R521G) mice escape early lethality; these escapers have modest motor impairments and altered sociability, which correspond with a reduction of dendritic arbors and mature spines. Remarkably, only FUS(R521G) mice show dendritic defects; FUS(WT) mice do not. Activation of metabotropic glutamate receptors 1/5 in neocortical slices and isolated synaptoneurosomes increases endogenous mouse FUS and FUS(WT) protein levels but decreases the FUS(R521G) protein, providing a potential biochemical basis for the dendritic spine differences between FUS(WT) and FUS(R521G) mice.
Collapse
|
93
|
De novo FUS mutations in 2 Korean patients with sporadic amyotrophic lateral sclerosis. Neurobiol Aging 2014; 36:1604.e17-9. [PMID: 25457557 DOI: 10.1016/j.neurobiolaging.2014.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/05/2014] [Indexed: 12/14/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disorder. Approximately 5% of ALS patients are familial (fALS) cases, and the remaining 95% are apparently sporadic (sALS) cases. To date, a number of genes have been discovered as associated with ALS, but the genetic background of sALS is not yet fully understood. The occurrence of de novo mutations in ALS genes might be an explanation for sALS, but reduced penetrance could be an alternative theory. Previously, we screened mutations in 5 ALS genes including SOD1 and FUS in 9 fALS and 249 sALS patients and found a total of 15 patients with either SOD1 (7 fALS and 3 sALS) or FUS (1 fALS and 4 sALS) mutations. Interestingly, only 1 fALS patient had the FUS mutation, whereas 4 sALS patients had mutations in this gene. To determine if the FUS mutations in sALS were de novo, we performed genetic analysis on 2 sALS patients with living parents. Genetic analysis confirmed that 2 FUS mutations, including the c.1483C>T (p.Arg495*) and the c.1509_1510delAG (p.Gly504Trpfs*12) mutations, were found only in the patients and not in their parents, confirming the de novo occurrence of these mutations. These findings support the notion that de novo mutations are responsible for a certain proportion of sALS.
Collapse
|
94
|
Tibshirani M, Tradewell ML, Mattina KR, Minotti S, Yang W, Zhou H, Strong MJ, Hayward LJ, Durham HD. Cytoplasmic sequestration of FUS/TLS associated with ALS alters histone marks through loss of nuclear protein arginine methyltransferase 1. Hum Mol Genet 2014; 24:773-86. [PMID: 25274782 DOI: 10.1093/hmg/ddu494] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mutations in the RNA-binding protein FUS/TLS (FUS) have been linked to the neurodegenerative disease amyotrophic lateral sclerosis (ALS). Although predominantly nuclear, this heterogenous nuclear ribonuclear protein (hnRNP) has multiple functions in RNA processing including intracellular trafficking. In ALS, mutant or wild-type (WT) FUS can form neuronal cytoplasmic inclusions. Asymmetric arginine methylation of FUS by the class 1 arginine methyltransferase, protein arginine methyltransferase 1 (PRMT1), regulates nucleocytoplasmic shuttling of FUS. In motor neurons of primary spinal cord cultures, redistribution of endogenous mouse and that of ectopically expressed WT or mutant human FUS to the cytoplasm led to nuclear depletion of PRMT1, abrogating methylation of its nuclear substrates. Specifically, hypomethylation of arginine 3 of histone 4 resulted in decreased acetylation of lysine 9/14 of histone 3 and transcriptional repression. Distribution of neuronal PRMT1 coincident with FUS also was detected in vivo in the spinal cord of FUS(R495X) transgenic mice. However, nuclear PRMT1 was not stable postmortem obviating meaningful evaluation of ALS autopsy cases. This study provides evidence for loss of PRMT1 function as a consequence of cytoplasmic accumulation of FUS in the pathogenesis of ALS, including changes in the histone code regulating gene transcription.
Collapse
Affiliation(s)
- Michael Tibshirani
- Montreal Neurological Institute and Department of Neurology/Neurosurgery, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Miranda L Tradewell
- Montreal Neurological Institute and Department of Neurology/Neurosurgery, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Katie R Mattina
- Montreal Neurological Institute and Department of Neurology/Neurosurgery, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Sandra Minotti
- Montreal Neurological Institute and Department of Neurology/Neurosurgery, McGill University, Montreal, Quebec, Canada H3A 2B4
| | - Wencheng Yang
- Robarts Research Institute, Western University, London, Ontario, Canada N6A 5C1 and
| | - Hongru Zhou
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Michael J Strong
- Robarts Research Institute, Western University, London, Ontario, Canada N6A 5C1 and
| | - Lawrence J Hayward
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Heather D Durham
- Montreal Neurological Institute and Department of Neurology/Neurosurgery, McGill University, Montreal, Quebec, Canada H3A 2B4
| |
Collapse
|
95
|
Calvo A, Moglia C, Canosa A, Brunetti M, Barberis M, Traynor BJ, Carrara G, Valentini C, Restagno G, Chiò A. De novo nonsense mutation of the FUS gene in an apparently familial amyotrophic lateral sclerosis case. Neurobiol Aging 2014; 35:1513.e7-11. [PMID: 24439481 PMCID: PMC3961545 DOI: 10.1016/j.neurobiolaging.2013.12.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 12/25/2013] [Indexed: 12/11/2022]
Abstract
Mutations in C9ORF72, SOD1, TARDBP, and FUS genes account for approximately two-third of familial cases and 5% of sporadic amyotrophic lateral sclerosis (ALS) cases. We present the first case of an ALS patient carrying a de novo nonsense mutation in exon 14 of the FUS gene (c.1483c>t; p.R495X) with an apparently familial ALS. This mutation causes a phenotype characterized by a young age at onset, a rapid course (<24 months), and a bulbar onset with early respiratory involvement with a predominant lower motor neuron disease. De novo mutations could account for a sizable number of apparently sporadic ALS patients carrying mutations of ALS-related genes.
Collapse
Affiliation(s)
- Andrea Calvo
- "Rita Levi Montalcini" Department of Neuroscience, ALS Center, University of Torino, Torino, Italy
| | - Cristina Moglia
- "Rita Levi Montalcini" Department of Neuroscience, ALS Center, University of Torino, Torino, Italy
| | - Antonio Canosa
- "Rita Levi Montalcini" Department of Neuroscience, ALS Center, University of Torino, Torino, Italy
| | - Maura Brunetti
- Laboratory of Molecular Genetics, Azienda Ospedaliera Città della Salute e della Scienza, Torino, Italy
| | - Marco Barberis
- Laboratory of Molecular Genetics, Azienda Ospedaliera Città della Salute e della Scienza, Torino, Italy
| | - Bryan J Traynor
- Neuromuscular Diseases Research Unit, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Giovanna Carrara
- Department of Neuroradiology, Azienda Ospedaliera Città della Salute e della Scienza, Torino, Italy
| | - Consuelo Valentini
- Department of Neuroradiology, Azienda Ospedaliera Città della Salute e della Scienza, Torino, Italy
| | - Gabriella Restagno
- Laboratory of Molecular Genetics, Azienda Ospedaliera Città della Salute e della Scienza, Torino, Italy
| | - Adriano Chiò
- "Rita Levi Montalcini" Department of Neuroscience, ALS Center, University of Torino, Torino, Italy; Neuroscience Institute of Torino, Torino, Italy.
| |
Collapse
|
96
|
Kenna KP, McLaughlin RL, Byrne S, Elamin M, Heverin M, Kenny EM, Cormican P, Morris DW, Donaghy CG, Bradley DG, Hardiman O. Delineating the genetic heterogeneity of ALS using targeted high-throughput sequencing. J Med Genet 2013; 50:776-83. [PMID: 23881933 PMCID: PMC3812897 DOI: 10.1136/jmedgenet-2013-101795] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Over 100 genes have been implicated in the aetiology of amyotrophic lateral sclerosis (ALS). A detailed understanding of their independent and cumulative contributions to disease burden may help guide various clinical and research efforts. METHODS Using targeted high-throughput sequencing, we characterised the variation of 10 Mendelian and 23 low penetrance/tentative ALS genes within a population-based cohort of 444 Irish ALS cases (50 fALS, 394 sALS) and 311 age-matched and geographically matched controls. RESULTS Known or potential high-penetrance ALS variants were identified within 17.1% of patients (38% of fALS, 14.5% of sALS). 12.8% carried variants of Mendelian disease genes (C9orf72 8.78%; SETX 2.48%; ALS2 1.58%; FUS 0.45%; TARDBP 0.45%; OPTN 0.23%; VCP 0.23%. ANG, SOD1, VAPB 0%), 4.7% carried variants of low penetrance/tentative ALS genes and 9.7% (30% of fALS, 7.1% of sALS) carried previously described ALS variants (C9orf72 8.78%; FUS 0.45%; TARDBP 0.45%). 1.6% of patients carried multiple known/potential disease variants, including all identified carriers of an established ALS variant (p<0.01); TARDBP:c.859G>A(p.[G287S]) (n=2/2 sALS). Comparison of our results with those from studies of other European populations revealed significant differences in the spectrum of disease variation (p=1.7×10(-4)). CONCLUSIONS Up to 17% of Irish ALS cases may carry high-penetrance variants within the investigated genes. However, the precise nature of genetic susceptibility differs significantly from that reported within other European populations. Certain variants may not cause disease in isolation and concomitant analysis of disease genes may prove highly important.
Collapse
Affiliation(s)
- Kevin P Kenna
- Smurfit Institute of Genetics, Trinity College, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Groen EJN, Fumoto K, Blokhuis AM, Engelen-Lee J, Zhou Y, van den Heuvel DMA, Koppers M, van Diggelen F, van Heest J, Demmers JAA, Kirby J, Shaw PJ, Aronica E, Spliet WGM, Veldink JH, van den Berg LH, Pasterkamp RJ. ALS-associated mutations in FUS disrupt the axonal distribution and function of SMN. Hum Mol Genet 2013; 22:3690-704. [PMID: 23681068 DOI: 10.1093/hmg/ddt222] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mutations in the RNA binding protein fused in sarcoma/translated in liposarcoma (FUS/TLS) cause amyotrophic lateral sclerosis (ALS). Although ALS-linked mutations in FUS often lead to a cytosolic mislocalization of the protein, the pathogenic mechanisms underlying these mutations remain poorly understood. To gain insight into these mechanisms, we examined the biochemical, cell biological and functional properties of mutant FUS in neurons. Expression of different FUS mutants (R521C, R521H, P525L) in neurons caused axonal defects. A protein interaction screen performed to explain these phenotypes identified numerous FUS interactors including the spinal muscular atrophy (SMA) causing protein survival motor neuron (SMN). Biochemical experiments showed that FUS and SMN interact directly and endogenously, and that this interaction can be regulated by FUS mutations. Immunostaining revealed co-localization of mutant FUS aggregates and SMN in primary neurons. This redistribution of SMN to cytosolic FUS accumulations led to a decrease in axonal SMN. Finally, cell biological experiments showed that overexpression of SMN rescued the axonal defects induced by mutant FUS, suggesting that FUS mutations cause axonal defects through SMN. This study shows that neuronal aggregates formed by mutant FUS protein may aberrantly sequester SMN and concomitantly cause a reduction of SMN levels in the axon, leading to axonal defects. These data provide a functional link between ALS-linked FUS mutations, SMN and neuronal connectivity and support the idea that different motor neuron disorders such as SMA and ALS may be caused, in part, by defects in shared molecular pathways.
Collapse
Affiliation(s)
- Ewout J N Groen
- Department of Neuroscience and Pharmacology, Rudolf Magnus Institute of Neuroscience, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
How do the RNA-binding proteins TDP-43 and FUS relate to amyotrophic lateral sclerosis and frontotemporal degeneration, and to each other? Curr Opin Neurol 2013; 25:701-7. [PMID: 23041957 DOI: 10.1097/wco.0b013e32835a269b] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review examines the recent research developments aimed at defining the role of RNA-binding proteins (TDP-43 and FUS) in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). RECENT FINDINGS TAR DNA-binding protein 43 kDa (TDP-43) and fused in sarcoma (FUS) are RNA-binding proteins that form aggregates in ALS and FTLD, and when mutated can drive the pathogenesis of these disorders. However, fundamental questions remain as to the relationship between TDP-43 and FUS aggregation and disease, their normal and pathologic function, and where they converge on the same cellular pathways. Autopsy series point to distinct molecular actions as TDP-43 and FUS neuronal inclusions do not overlap, with FUS inclusions being present in only a small subgroup of patients. By contrast, modeling experiments in lower organisms support a genetic interaction between TDP-43 and FUS, although it is likely indirect. Regardless, the recent finding that additional RNA-binding proteins may also cause ALS, and the observation that TDP-43 aggregation remains a core feature in all of the recently identified genetic forms of ALS (C9ORF72, VCP, UBQLN2, and PFN1), underscores the central role of TDP-43 and RNA metabolism in ALS and FTLD. SUMMARY Recent discoveries point to an unprecedented convergence of molecular pathways in ALS and FTLD involving RNA metabolism. Defining the exact points of convergence will likely be key to advancing therapeutics development in the coming years.
Collapse
|
99
|
Lattante S, Rouleau GA, Kabashi E. TARDBPandFUSMutations Associated with Amyotrophic Lateral Sclerosis: Summary and Update. Hum Mutat 2013; 34:812-26. [DOI: 10.1002/humu.22319] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 02/28/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Serena Lattante
- Institut du Cerveau et de la Moelle épinière; Centre de Recherche, CHU Pitié-Salpétrière, Inserm, UMR_S975, CRICM, F-75013; UPMC Univ Paris 06, UMR_S975, F-75013; CNRS UMR 7225; F-75013; Paris; France
| | - Guy A. Rouleau
- Montreal Neurological Institute; Department of Neurology and Neurosurgery, McGill University; Montreal; Canada
| | | |
Collapse
|
100
|
Sabatelli M, Conte A, Zollino M. Clinical and genetic heterogeneity of amyotrophic lateral sclerosis. Clin Genet 2013; 83:408-16. [DOI: 10.1111/cge.12117] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 01/30/2013] [Accepted: 01/30/2013] [Indexed: 02/01/2023]
Affiliation(s)
| | | | - M Zollino
- Istituto di Genetica Medica; Università Cattolica del Sacro Cuore; Rome; Italy
| |
Collapse
|