51
|
Alzheimer’s Disease and Frontotemporal Lobar Degeneration: Mouse Models. NEURODEGENER DIS 2018. [DOI: 10.1007/978-3-319-72938-1_10] [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
|
52
|
Serpente M, Galimberti D. Autosomal Dominant Frontotemporal Lobar Degeneration: From Genotype to Phenotype. NEURODEGENER DIS 2018. [DOI: 10.1007/978-3-319-72938-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
53
|
Goutman SA, Chen KS, Paez-Colasante X, Feldman EL. Emerging understanding of the genotype-phenotype relationship in amyotrophic lateral sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2018; 148:603-623. [PMID: 29478603 DOI: 10.1016/b978-0-444-64076-5.00039-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, noncurable neurodegenerative disorder of the upper and lower motor neurons causing weakness and death within a few years of symptom onset. About 10% of patients with ALS have a family history of the disease; however, ALS-associated genetic mutations are also found in sporadic cases. There are over 100 ALS-associated mutations, and importantly, several genetic mutations, including C9ORF72, SOD1, and TARDBP, have led to mechanistic insight into this complex disease. In the clinical realm, knowledge of ALS genetics can also help explain phenotypic heterogeneity, aid in genetic counseling, and in the future may help direct treatment efforts.
Collapse
Affiliation(s)
- Stephen A Goutman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States.
| | - Kevin S Chen
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | | | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
54
|
Clayton EL, Mancuso R, Nielsen TT, Mizielinska S, Holmes H, Powell N, Norona F, Larsen JO, Milioto C, Wilson KM, Lythgoe MF, Ourselin S, Nielsen JE, Johannsen P, Holm I, Collinge J, Oliver PL, Gomez-Nicola D, Isaacs AM. Early microgliosis precedes neuronal loss and behavioural impairment in mice with a frontotemporal dementia-causing CHMP2B mutation. Hum Mol Genet 2017; 26:873-887. [PMID: 28093491 PMCID: PMC5409096 DOI: 10.1093/hmg/ddx003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/03/2017] [Indexed: 01/13/2023] Open
Abstract
Frontotemporal dementia (FTD)-causing mutations in the CHMP2B gene lead to the generation of mutant C-terminally truncated CHMP2B. We report that transgenic mice expressing endogenous levels of mutant CHMP2B developed late-onset brain volume loss associated with frank neuronal loss and FTD-like changes in social behaviour. These data are the first to show neurodegeneration in mice expressing mutant CHMP2B and indicate that our mouse model is able to recapitulate neurodegenerative changes observed in FTD. Neuroinflammation has been increasingly implicated in neurodegeneration, including FTD. Therefore, we investigated neuroinflammation in our CHMP2B mutant mice. We observed very early microglial proliferation that develops into a clear pro-inflammatory phenotype at late stages. Importantly, we also observed a similar inflammatory profile in CHMP2B patient frontal cortex. Aberrant microglial function has also been implicated in FTD caused by GRN, MAPT and C9orf72 mutations. The presence of early microglial changes in our CHMP2B mutant mice indicates neuroinflammation may be a contributing factor to the neurodegeneration observed in FTD.
Collapse
Affiliation(s)
- Emma L Clayton
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Renzo Mancuso
- Biological Sciences, University of Southampton, Southampton General Hospital, South Laboratory and Pathology Block, Tremona Road, Southampton SO166YD, UK
| | - Troels Tolstrup Nielsen
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Denmark
| | - Sarah Mizielinska
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Holly Holmes
- Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Nicholas Powell
- Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, 72 Huntley Street, London WC1E 6DD, UK.,Faculty of Health and Medical Sciences, Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Frances Norona
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Jytte Overgaard Larsen
- Translational Imaging Group, Centre for Medical Image Computing (CMIC), University College London, UK
| | - Carmelo Milioto
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Katherine M Wilson
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Mark F Lythgoe
- Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Sebastian Ourselin
- Faculty of Health and Medical Sciences, Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jörgen E Nielsen
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Denmark.,Section of Neurogenetics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Peter Johannsen
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Denmark
| | - Ida Holm
- Laboratory for Experimental Neuropathology, Department of Pathology, Randers Hospital, DK-8930 Randers NØ, Denmark.,Institute of Clinical Medicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - John Collinge
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.,MRC Prion Unit, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | | | - Peter L Oliver
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Diego Gomez-Nicola
- Biological Sciences, University of Southampton, Southampton General Hospital, South Laboratory and Pathology Block, Tremona Road, Southampton SO166YD, UK
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
55
|
Gao FB, Almeida S, Lopez-Gonzalez R. Dysregulated molecular pathways in amyotrophic lateral sclerosis-frontotemporal dementia spectrum disorder. EMBO J 2017; 36:2931-2950. [PMID: 28916614 DOI: 10.15252/embj.201797568] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/15/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia (FTD), the second most common form of dementia in people under 65 years of age, is characterized by progressive atrophy of the frontal and/or temporal lobes. FTD overlaps extensively with the motor neuron disease amyotrophic lateral sclerosis (ALS), especially at the genetic level. Both FTD and ALS can be caused by many mutations in the same set of genes; the most prevalent of these mutations is a GGGGCC repeat expansion in the first intron of C9ORF72 As shown by recent intensive studies, some key cellular pathways are dysregulated in the ALS-FTD spectrum disorder, including autophagy, nucleocytoplasmic transport, DNA damage repair, pre-mRNA splicing, stress granule dynamics, and others. These exciting advances reveal the complexity of the pathogenic mechanisms of FTD and ALS and suggest promising molecular targets for future therapeutic interventions in these devastating disorders.
Collapse
Affiliation(s)
- Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | | |
Collapse
|
56
|
De Vos KJ, Hafezparast M. Neurobiology of axonal transport defects in motor neuron diseases: Opportunities for translational research? Neurobiol Dis 2017; 105:283-299. [PMID: 28235672 PMCID: PMC5536153 DOI: 10.1016/j.nbd.2017.02.004] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/26/2017] [Accepted: 02/20/2017] [Indexed: 12/12/2022] Open
Abstract
Intracellular trafficking of cargoes is an essential process to maintain the structure and function of all mammalian cell types, but especially of neurons because of their extreme axon/dendrite polarisation. Axonal transport mediates the movement of cargoes such as proteins, mRNA, lipids, membrane-bound vesicles and organelles that are mostly synthesised in the cell body and in doing so is responsible for their correct spatiotemporal distribution in the axon, for example at specialised sites such as nodes of Ranvier and synaptic terminals. In addition, axonal transport maintains the essential long-distance communication between the cell body and synaptic terminals that allows neurons to react to their surroundings via trafficking of for example signalling endosomes. Axonal transport defects are a common observation in a variety of neurodegenerative diseases, and mutations in components of the axonal transport machinery have unequivocally shown that impaired axonal transport can cause neurodegeneration (reviewed in El-Kadi et al., 2007, De Vos et al., 2008; Millecamps and Julien, 2013). Here we review our current understanding of axonal transport defects and the role they play in motor neuron diseases (MNDs) with a specific focus on the most common form of MND, amyotrophic lateral sclerosis (ALS).
Collapse
Affiliation(s)
- Kurt J De Vos
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Majid Hafezparast
- Neuroscience, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK.
| |
Collapse
|
57
|
Sadoul R, Laporte MH, Chassefeyre R, Chi KI, Goldberg Y, Chatellard C, Hemming FJ, Fraboulet S. The role of ESCRT during development and functioning of the nervous system. Semin Cell Dev Biol 2017; 74:40-49. [PMID: 28811263 DOI: 10.1016/j.semcdb.2017.08.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/21/2017] [Accepted: 08/04/2017] [Indexed: 12/12/2022]
Abstract
The endosomal sorting complex required for transport (ESCRT) is made of subcomplexes (ESCRT 0-III), crucial to membrane remodelling at endosomes, nuclear envelope and cell surface. ESCRT-III shapes membranes and in most cases cooperates with the ATPase VPS4 to mediate fission of membrane necks from the inside. The first ESCRT complexes mainly serve to catalyse the formation of ESCRT-III but can be bypassed by accessory proteins like the Alg-2 interacting protein-X (ALIX). In the nervous system, ALIX/ESCRT controls the survival of embryonic neural progenitors and later on the outgrowth and pruning of axons and dendrites, all necessary steps to establish a functional brain. In the adult brain, ESCRTs allow the endosomal turn over of synaptic vesicle proteins while stable ESCRT complexes might serve as scaffolds for the postsynaptic parts. The necessity of ESCRT for the harmonious function of the brain has its pathological counterpart, the mutations in CHMP2B of ESCRT-III giving rise to several neurodegenerative diseases.
Collapse
Affiliation(s)
- Rémy Sadoul
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France.
| | - Marine H Laporte
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Romain Chassefeyre
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Kwang Il Chi
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Yves Goldberg
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Christine Chatellard
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Fiona J Hemming
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Sandrine Fraboulet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| |
Collapse
|
58
|
Rostgaard N, Roos P, Budtz-Jørgensen E, Johannsen P, Waldemar G, Nørremølle A, Lindquist SG, Gydesen S, Brown JM, Collinge J, Isaacs AM, Nielsen TT, Nielsen JE. TMEM106B and ApoE polymorphisms in CHMP2B-mediated frontotemporal dementia (FTD-3). Neurobiol Aging 2017; 59:221.e1-221.e7. [PMID: 28888721 DOI: 10.1016/j.neurobiolaging.2017.06.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 06/24/2017] [Accepted: 06/30/2017] [Indexed: 12/12/2022]
Abstract
Single-nucleotide polymorphisms in the TMEM106B gene have been identified as a risk factor in frontotemporal dementia (FTD). The major allele of SNP rs3173615 is a risk factor in sporadic FTD, whereas the minor allele seems protective in GRN- and C9orf72-mediated FTD. The role of apolipoprotein E (ApoE) in FTD is uncertain, though an established risk factor in Alzheimer's disease. In a unique Danish family, inherited FTD is caused by a mutation in the CHMP2B gene located on chromosome 3 (FTD-3). In this family, both risk factors TMEM106B and ApoE were analyzed and correlated to age at onset (AAO) and progression in terms of age at institutionalization (AAI) and age at death (AAD). Although TMEM106B and CHMP2B share cellular function in that both localize to endolysosomes, TMEM106B genotypes appeared to have no influence on the clinical disease course. ApoE ε4 was found to be a protective factor with later AAO and AAI, whereas ε2 seemed to aggravate the disease with earlier AAO and AAD. These results indicate ApoE ε2 as a risk factor in FTD-3 and suggest a protective role of ε4.
Collapse
Affiliation(s)
- Nina Rostgaard
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Section 6911, Copenhagen, Denmark
| | - Peter Roos
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Section 6911, Copenhagen, Denmark.
| | | | - Peter Johannsen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Section 6911, Copenhagen, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Section 6911, Copenhagen, Denmark
| | - Anne Nørremølle
- Section of Neurogenetics, Institute of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| | - Suzanne G Lindquist
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Section 6911, Copenhagen, Denmark; Department of Clinical Genetics, Rigshospitalet, University of Copenhagen, Section 4062, Copenhagen, Denmark
| | - Susanne Gydesen
- Department of Psychiatry, Community Mental Health Services in Region Zealand, Nykøbing Sjælland, Denmark
| | - Jeremy M Brown
- Department of Neurology, Addenbrooke's Hospital, Cambridge, UK
| | - John Collinge
- MRC Prion Unit, Institute of Neurology, London, UK; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, UCL Institute of Neurology, London, UK
| | | | - Troels T Nielsen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Section 6911, Copenhagen, Denmark
| | - Jørgen E Nielsen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Section 6911, Copenhagen, Denmark; Section of Neurogenetics, Institute of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
59
|
Pottier C, Ravenscroft TA, Sanchez-Contreras M, Rademakers R. Genetics of FTLD: overview and what else we can expect from genetic studies. J Neurochem 2017; 138 Suppl 1:32-53. [PMID: 27009575 DOI: 10.1111/jnc.13622] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/26/2016] [Accepted: 03/18/2016] [Indexed: 12/11/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) comprises a highly heterogeneous group of disorders clinically associated with behavioral and personality changes, language impairment, and deficits in executive functioning, and pathologically associated with degeneration of frontal and temporal lobes. Some patients present with motor symptoms including amyotrophic lateral sclerosis. Genetic research over the past two decades in FTLD families led to the identification of three common FTLD genes (microtubule-associated protein tau, progranulin, and chromosome 9 open reading frame 72) and a small number of rare FTLD genes, explaining the disease in almost all autosomal dominant FTLD families but only a minority of apparently sporadic patients or patients in whom the family history is less clear. Identification of additional FTLD (risk) genes is therefore highly anticipated, especially with the emerging use of next-generation sequencing. Common variants in the transmembrane protein 106 B were identified as a genetic risk factor of FTLD and disease modifier in patients with known mutations. This review summarizes for each FTLD gene what we know about the type and frequency of mutations, their associated clinical and pathological features, and potential disease mechanisms. We also provide an overview of emerging disease pathways encompassing multiple FTLD genes. We further discuss how FTLD specific issues, such as disease heterogeneity, the presence of an unclear family history and the possible role of an oligogenic basis of FTLD, can pose challenges for future FTLD gene identification and risk assessment of specific variants. Finally, we highlight emerging clinical, genetic, and translational research opportunities that lie ahead. Genetic research led to the identification of three common FTLD genes with rare variants (MAPT, GRN, and C9orf72) and a small number of rare genes. Efforts are now ongoing, which aimed at the identification of rare variants with high risk and/or low frequency variants with intermediate effect. Common risk variants have also been identified, such as TMEM106B. This review discusses the current knowledge on FTLD genes and the emerging disease pathways encompassing multiple FTLD genes.
Collapse
Affiliation(s)
- Cyril Pottier
- Mayo Clinic Jacksonville, Department of Neuroscience, Jacksonville, FL, USA
| | | | | | - Rosa Rademakers
- Mayo Clinic Jacksonville, Department of Neuroscience, Jacksonville, FL, USA
| |
Collapse
|
60
|
Zhang Y, Schmid B, Nikolaisen NK, Rasmussen MA, Aldana BI, Agger M, Calloe K, Stummann TC, Larsen HM, Nielsen TT, Huang J, Xu F, Liu X, Bolund L, Meyer M, Bak LK, Waagepetersen HS, Luo Y, Nielsen JE, Holst B, Clausen C, Hyttel P, Freude KK. Patient iPSC-Derived Neurons for Disease Modeling of Frontotemporal Dementia with Mutation in CHMP2B. Stem Cell Reports 2017; 8:648-658. [PMID: 28216144 PMCID: PMC5355623 DOI: 10.1016/j.stemcr.2017.01.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 12/23/2022] Open
Abstract
The truncated mutant form of the charged multivesicular body protein 2B (CHMP2B) is causative for frontotemporal dementia linked to chromosome 3 (FTD3). CHMP2B is a constituent of the endosomal sorting complex required for transport (ESCRT) and, when mutated, disrupts endosome-to-lysosome trafficking and substrate degradation. To understand the underlying molecular pathology, FTD3 patient induced pluripotent stem cells (iPSCs) were differentiated into forebrain-type cortical neurons. FTD3 neurons exhibited abnormal endosomes, as previously shown in patients. Moreover, mitochondria of FTD3 neurons displayed defective cristae formation, accompanied by deficiencies in mitochondrial respiration and increased levels of reactive oxygen. In addition, we provide evidence for perturbed iron homeostasis, presenting an in vitro patient-specific model to study the effects of iron accumulation in neurodegenerative diseases. All phenotypes observed in FTD3 neurons were rescued in CRISPR/Cas9-edited isogenic controls. These findings illustrate the relevance of our patient-specific in vitro models and open up possibilities for drug target development. FTD3 neurons show abnormalities in endosomes and mitochondria Parkinson's and Alzheimer's disease core genes are altered in FTD3 neurons Iron homeostasis is perturbed in FTD3 neurons Impairments in FTD3 neurons are rescued in CRISPR/Cas9-edited isogenic controls
Collapse
Affiliation(s)
- Yu Zhang
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark.
| | | | - Nanett K Nikolaisen
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | | | - Blanca I Aldana
- Neurometabolism Research Unit, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Mikkel Agger
- Stem Cell and Developmental Neurobiology Group, Department of Neurobiology Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Kirstine Calloe
- The Physiology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | | | - Hjalte M Larsen
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - Troels T Nielsen
- Neurogenetics Clinic & Research Lab, Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jinrong Huang
- BGI-Shenzhen, 518083 Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China
| | - Fengping Xu
- BGI-Shenzhen, 518083 Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China
| | - Xin Liu
- BGI-Shenzhen, 518083 Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China
| | - Lars Bolund
- Danish Regenerative Engineering Alliance for Medicine (DREAM), Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Morten Meyer
- Stem Cell and Developmental Neurobiology Group, Department of Neurobiology Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Lasse K Bak
- Neurometabolism Research Unit, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Helle S Waagepetersen
- Neurometabolism Research Unit, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Yonglun Luo
- Danish Regenerative Engineering Alliance for Medicine (DREAM), Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Jørgen E Nielsen
- Neurogenetics Clinic & Research Lab, Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | | | | | | | - Poul Hyttel
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark
| | - Kristine K Freude
- Stem Cells and Embryology Group, Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg C, Denmark.
| |
Collapse
|
61
|
Tan RH, Ke YD, Ittner LM, Halliday GM. ALS/FTLD: experimental models and reality. Acta Neuropathol 2017; 133:177-196. [PMID: 28058507 DOI: 10.1007/s00401-016-1666-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/19/2016] [Accepted: 12/30/2016] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis is characterised by a loss of upper and lower motor neurons and characteristic muscle weakness and wasting, the most common form being sporadic disease with neuronal inclusions containing the tar DNA-binding protein 43 (TDP-43). Frontotemporal lobar degeneration is characterised by atrophy of the frontal and/or temporal lobes, the most common clinical form being the behavioural variant, in which neuronal inclusions containing either TDP-43 or 3-repeat tau are most prevalent. Although the genetic mutations associated with these diseases have allowed various experimental models to be developed, the initial genetic forms identified remain the most common models employed to date. It is now known that these first models faithfully recapitulate only some aspects of these diseases and do not represent the majority of cases or the most common overlapping pathologies. Newer models targeting the main molecular pathologies are still rare and in some instances, lack significant aspects of the molecular pathology. However, these diseases are complex and multigenic, indicating that experimental models may need to be targeted to different disease aspects. This would allow information to be gleaned from a variety of different yet relevant models, each of which has the capacity to capture a certain aspect of the disease, and together will enable a more complete understanding of these complex and multi-layered diseases.
Collapse
Affiliation(s)
- Rachel H Tan
- Neuroscience Research Australia, Randwick, NSW, 2031, Australia
- School of Medical Sciences, University of NSW, Sydney, NSW, 2052, Australia
- Brain and Mind Centre, Sydney Medical School, the University of Sydney, Sydney, NSW, 2006, Australia
| | - Yazi D Ke
- Motor Neuron Disease Unit, Department of Anatomy, Faculty of Medicine, University of NSW, Sydney, NSW, 2052, Australia
| | - Lars M Ittner
- Neuroscience Research Australia, Randwick, NSW, 2031, Australia.
- Dementia Research Unit, Department of Anatomy, Faculty of Medicine, University of NSW, Sydney, NSW, 2052, Australia.
| | - Glenda M Halliday
- Neuroscience Research Australia, Randwick, NSW, 2031, Australia.
- School of Medical Sciences, University of NSW, Sydney, NSW, 2052, Australia.
- Brain and Mind Centre, Sydney Medical School, the University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
62
|
Götzl JK, Lang CM, Haass C, Capell A. Impaired protein degradation in FTLD and related disorders. Ageing Res Rev 2016; 32:122-139. [PMID: 27166223 DOI: 10.1016/j.arr.2016.04.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/21/2016] [Accepted: 04/23/2016] [Indexed: 12/12/2022]
Abstract
Impaired protein degradation has been discussed as a cause or consequence of various neurodegenerative diseases, such as Alzheimer's, Parkinson's and Huntington's disease. More recently, evidence accumulated that dysfunctional protein degradation may play a role in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Since in almost all neurodegenerative diseases, protein aggregates are disease-defining hallmarks, it is most likely that impaired protein degradation contributes to disease onset and progression. In the majority of FTD cases, the pathological protein aggregates contain either microtubuleassociated protein tau or TAR DNA-binding protein (TDP)-43. Aggregates are also positive for ubiquitin and p62/sequestosome 1 (SQSTM1) indicating that these aggregates are targeted for degradation. FTD-linked mutations in genes encoding three autophagy adaptor proteins, p62/SQSTM1, ubiquilin 2 and optineurin, indicate that impaired autophagy might cause FTD. Furthermore, the strongest evidence for lysosomal impairment in FTD is provided by the progranulin (GRN) gene, which is linked to FTD and neuronal ceroid lipofuscinosis. In this review, we summarize the observations that have been made during the last years linking the accumulation of disease-associated proteins in FTD to impaired protein degradation pathways. In addition, we take resent findings for nucleocytoplasmic transport defects of TDP-43, as discussed for hexanucleotide repeat expansions in C9orf72 into account and provide a hypothesis how the interplay of altered nuclear transport and protein degradation leads to the accumulation of protein deposits.
Collapse
|
63
|
Vernay A, Therreau L, Blot B, Risson V, Dirrig-Grosch S, Waegaert R, Lequeu T, Sellal F, Schaeffer L, Sadoul R, Loeffler JP, René F. A transgenic mouse expressing CHMP2Bintron5 mutant in neurons develops histological and behavioural features of amyotrophic lateral sclerosis and frontotemporal dementia. Hum Mol Genet 2016; 25:3341-3360. [PMID: 27329763 DOI: 10.1093/hmg/ddw182] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022] Open
Abstract
Mutations in the charged multivesicular body protein 2B (CHMP2B) are associated with frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS), and with a mixed ALS-FTD syndrome. To model this syndrome, we generated a transgenic mouse line expressing the human CHMP2Bintron5 mutant in a neuron-specific manner. These mice developed a dose-dependent disease phenotype. A longitudinal study revealed progressive gait abnormalities, reduced muscle strength and decreased motor coordination. CHMP2Bintron5 mice died due to generalized paralysis. When paralyzed, signs of denervation were present as attested by altered electromyographic profiles, by decreased number of fully innervated neuromuscular junctions, by reduction in size of motor endplates and by a decrease of sciatic nerve axons area. However, spinal motor neurons cell bodies were preserved until death. In addition to the motor dysfunctions, CHMP2Bintron5 mice progressively developed FTD-relevant behavioural modifications such as disinhibition, stereotypies, decrease in social interactions, compulsivity and change in dietary preferences. Furthermore, neurons in the affected spinal cord and brain regions showed accumulation of p62-positive cytoplasmic inclusions associated or not with ubiquitin and CHMP2Bintron5 As observed in FTD3 patients, these inclusions were negative for TDP-43 and FUS. Moreover, astrogliosis and microgliosis developed with age. Altogether, these data indicate that the neuronal expression of human CHMP2Bintron5 in areas involved in motor and cognitive functions induces progressive motor alterations associated with dementia symptoms and with histopathological hallmarks reminiscent of both ALS and FTD.
Collapse
Affiliation(s)
- Aurélia Vernay
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Ludivine Therreau
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Béatrice Blot
- INSERM U836, Grenoble Institut des Neurosciences, Université Joseph Fourier, F-38700 La Tronche, France
| | - Valérie Risson
- Laboratoire de Biologie Moléculaire de la Cellule, UMR5239 CNRS/ENS Lyon/UCBL/HCL Ecole normale supérieure de Lyon, F-69364 Lyon Cedex 07, France
| | - Sylvie Dirrig-Grosch
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Robin Waegaert
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Thiebault Lequeu
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - François Sellal
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Neurology department, Hôpitaux civils and CMRR, F-68000 Colmar, France
| | - Laurent Schaeffer
- Laboratoire de Biologie Moléculaire de la Cellule, UMR5239 CNRS/ENS Lyon/UCBL/HCL Ecole normale supérieure de Lyon, F-69364 Lyon Cedex 07, France
| | - Rémy Sadoul
- INSERM U836, Grenoble Institut des Neurosciences, Université Joseph Fourier, F-38700 La Tronche, France
| | - Jean-Philippe Loeffler
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Frédérique René
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France .,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| |
Collapse
|
64
|
Denora PS, Smets K, Zolfanelli F, Ceuterick-de Groote C, Casali C, Deconinck T, Sieben A, Gonzales M, Zuchner S, Darios F, Peeters D, Brice A, Malandrini A, De Jonghe P, Santorelli FM, Stevanin G, Martin JJ, El Hachimi KH. Motor neuron degeneration in spastic paraplegia 11 mimics amyotrophic lateral sclerosis lesions. Brain 2016; 139:1723-34. [PMID: 27016404 PMCID: PMC5839621 DOI: 10.1093/brain/aww061] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/31/2016] [Indexed: 12/12/2022] Open
Abstract
The most common form of autosomal recessive hereditary spastic paraplegia is caused by
mutations in the SPG11/KIAA1840 gene on chromosome 15q.
The nature of the vast majority of SPG11 mutations found to date suggests
a loss-of-function mechanism of the encoded protein, spatacsin. The SPG11 phenotype is, in
most cases, characterized by a progressive spasticity with neuropathy, cognitive
impairment and a thin corpus callosum on brain MRI. Full neuropathological
characterization has not been reported to date despite the description of >100
SPG11 mutations. We describe here the clinical and pathological
features observed in two unrelated females, members of genetically ascertained SPG11
families originating from Belgium and Italy, respectively. We confirm the presence of
lesions of motor tracts in medulla oblongata and spinal cord associated with other lesions
of the central nervous system. Interestingly, we report for the first time pathological
hallmarks of SPG11 in neurons that include intracytoplasmic granular lysosome-like
structures mainly in supratentorial areas, and others in subtentorial areas that are
partially reminiscent of those observed in amyotrophic lateral sclerosis, such as
ubiquitin and p62 aggregates, except that they are never labelled with anti-TDP-43 or
anti-cystatin C. The neuropathological overlap with amyotrophic lateral sclerosis,
associated with some shared clinical manifestations, opens up new fields of investigation
in the physiopathological continuum of motor neuron degeneration.
Collapse
Affiliation(s)
- Paola S Denora
- 1 Ecole Pratique des Hautes Etudes, EPHE, PSL université, laboratoire de neurogénétique, F-75013, Paris, France 2 Inserm, U1127, F-75013, Paris, France 3 CNRS, UMR7225, F-75013, Paris, France 4 Sorbonne Universités, UPMC Univ Paris 06, UMR_S1127, Institut du Cerveau et de la Moelle épinière - ICM, Pitié-Salpêtrière Hospital, F-75013, Paris, France 5 Department of Genetics and Rare Diseases, IRCCS Bambino Gesu' Children Hospital, Rome, Italy
| | - Katrien Smets
- 6 Neurogenetics Group, VIB-Department of Molecular Genetics, University of Antwerp, Belgium 7 Laboratories of Neurogenetics, Institute Born-Bunge, University of Antwerp, Belgium 8 Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | | | | | - Carlo Casali
- 11 Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Polo Pontino Rome, Italy
| | - Tine Deconinck
- 6 Neurogenetics Group, VIB-Department of Molecular Genetics, University of Antwerp, Belgium 7 Laboratories of Neurogenetics, Institute Born-Bunge, University of Antwerp, Belgium
| | - Anne Sieben
- 10 Institute Born-Bunge, University of Antwerp, Belgium 12 Department of Neurology, University Hospital Gent, Belgium
| | - Michael Gonzales
- 13 Department of Human Genetics and Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Stephan Zuchner
- 13 Department of Human Genetics and Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Frédéric Darios
- 2 Inserm, U1127, F-75013, Paris, France 3 CNRS, UMR7225, F-75013, Paris, France 4 Sorbonne Universités, UPMC Univ Paris 06, UMR_S1127, Institut du Cerveau et de la Moelle épinière - ICM, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | - Dirk Peeters
- 14 Department of Neurology, AZ Groeninge, Kortrijk, Belgium
| | - Alexis Brice
- 2 Inserm, U1127, F-75013, Paris, France 3 CNRS, UMR7225, F-75013, Paris, France 4 Sorbonne Universités, UPMC Univ Paris 06, UMR_S1127, Institut du Cerveau et de la Moelle épinière - ICM, Pitié-Salpêtrière Hospital, F-75013, Paris, France 15 APHP, Département de Génétique, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | - Alessandro Malandrini
- 16 Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Peter De Jonghe
- 6 Neurogenetics Group, VIB-Department of Molecular Genetics, University of Antwerp, Belgium 7 Laboratories of Neurogenetics, Institute Born-Bunge, University of Antwerp, Belgium 8 Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Filippo M Santorelli
- 17 Molecular Medicine Laboratory, IRCCS Stella Maris Foundation, Calambrone, Pisa, Italy
| | - Giovanni Stevanin
- 1 Ecole Pratique des Hautes Etudes, EPHE, PSL université, laboratoire de neurogénétique, F-75013, Paris, France 2 Inserm, U1127, F-75013, Paris, France 3 CNRS, UMR7225, F-75013, Paris, France 4 Sorbonne Universités, UPMC Univ Paris 06, UMR_S1127, Institut du Cerveau et de la Moelle épinière - ICM, Pitié-Salpêtrière Hospital, F-75013, Paris, France 15 APHP, Département de Génétique, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | | | - Khalid H El Hachimi
- 1 Ecole Pratique des Hautes Etudes, EPHE, PSL université, laboratoire de neurogénétique, F-75013, Paris, France 2 Inserm, U1127, F-75013, Paris, France 3 CNRS, UMR7225, F-75013, Paris, France 4 Sorbonne Universités, UPMC Univ Paris 06, UMR_S1127, Institut du Cerveau et de la Moelle épinière - ICM, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| |
Collapse
|
65
|
Krasniak CS, Ahmad ST. The role of CHMP2B Intron5 in autophagy and frontotemporal dementia. Brain Res 2016; 1649:151-157. [PMID: 26972529 DOI: 10.1016/j.brainres.2016.02.051] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 02/05/2016] [Accepted: 02/10/2016] [Indexed: 12/12/2022]
Abstract
Charged multivesicular body protein 2B (CHMP2B) - a component of the endosomal complex required for transport-III (ESCRT-III) - is responsible for the vital membrane deformation functions in autophagy and endolysosomal trafficking. A dominant mutation in CHMP2B (CHMP2BIntron5) is associated with a subset of heritable frontotemporal dementia - frontotemporal dementia linked to chromosome 3 (FTD-3). ESCRT-III recruits Vps4, an AAA-ATPase that abscises the membrane during various cellular processes including autophagy and intraluminal vesicle formation. CHMP2BIntron5 results in a C-terminus truncation removing an important Vps4 binding site as well as eliminating the normal autoinhibitory resting state of CHMP2B. CHMP2B is expressed in most cell types but seems to be especially vital for proper neuronal function. CHMP2BIntron5-mediated phenotypes include misregulation of transmembrane receptors, accumulation of multilamellar structures, abnormal lysosomal morphology, down regulation of a brain-specific micro RNA (miRNA-124), abnormal dendritic spine morphology, decrease in dendritic arborization, and cell death. Currently, transgenic-fly,-mouse, and -human cell lines are being used to better understand the diverse phenotypes and develop therapeutic approaches for the CHMP2BIntron5-induced FTD-3. This article is part of a Special Issue entitled SI:Autophagy.
Collapse
Affiliation(s)
| | - S Tariq Ahmad
- Department of Biology, Colby College, 5720 Mayflower Hill, Waterville, ME 04901, USA.
| |
Collapse
|
66
|
Spencer B, Kim C, Gonzalez T, Bisquertt A, Patrick C, Rockenstein E, Adame A, Lee SJ, Desplats P, Masliah E. α-Synuclein interferes with the ESCRT-III complex contributing to the pathogenesis of Lewy body disease. Hum Mol Genet 2016; 25:1100-15. [PMID: 26740557 DOI: 10.1093/hmg/ddv633] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/30/2015] [Indexed: 01/17/2023] Open
Abstract
α-Synuclein (α-syn) has been implicated in neurological disorders with parkinsonism, including Parkinson's disease and Dementia with Lewy body. Recent studies have shown α-syn oligomers released from neurons can propagate from cell-to-cell in a prion-like fashion exacerbating neurodegeneration. In this study, we examined the role of the endosomal sorting complex required for transport (ESCRT) pathway on the propagation of α-syn. α-syn, which is transported via the ESCRT pathway through multivesicular bodies for degradation, can also target the degradation of the ESCRT protein-charged multivesicular body protein (CHMP2B), thus generating a roadblock of endocytosed α-syn. Disruption of the ESCRT transport system also resulted in increased exocytosis of α-syn thus potentially increasing cell-to-cell propagation of synuclein. Conversely, delivery of a lentiviral vector overexpressing CHMP2B rescued the neurodegeneration in α-syn transgenic mice. Better understanding of the mechanisms of intracellular trafficking of α-syn might be important for understanding the pathogenesis and developing new treatments for synucleinopathies.
Collapse
Affiliation(s)
| | - Changyoun Kim
- Department of Neuroscience and Department of Medicine, College of Medicine, Seoul National University, Seoul 110-799, Korea
| | | | | | | | | | | | - Seung-Jae Lee
- Department of Medicine, College of Medicine, Seoul National University, Seoul 110-799, Korea
| | | | - Eliezer Masliah
- Department of Neuroscience and Department of Pathology, University of California, San Diego, San Diego, CA 92103, USA and
| |
Collapse
|
67
|
Blocking IGF Signaling in Adult Neurons Alleviates Alzheimer's Disease Pathology through Amyloid-β Clearance. J Neurosci 2015; 35:11500-13. [PMID: 26290229 DOI: 10.1523/jneurosci.0343-15.2015] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Alzheimer's disease (AD) is a frequent and irreversible age-related neurodegeneration without efficient treatment. Experimental AD in mice responds positively to decreased insulin-like growth factor I (IGF-I) signaling, a pathway also implicated in aging. Here we aimed to protect the aging brain from devastating amyloid pathology by making specifically adult neurons resistant to IGF signaling. To achieve that, we knocked out neuronal IGF-1R during adulthood in APP/PS1 mice. We found that mutants exhibited improved spatial memory and reduced anxiety. Mutant brains displayed fewer amyloid plaques, less amyloid-β (Aβ), and diminished neuroinflammation. Surprisingly, adult neurons undergoing IGF-1R knock-out reduced their apical soma and developed leaner dendrites, indicative of remarkable structural plasticity entailing condensed forebrain neuroarchitecture. Neurons lacking IGF-1R in AD showed less accumulation of Aβ-containing autophagic vacuoles. At the same time, plasma Aβ levels were increased. Our data indicate that neuronal IGF-1R ablation, via preserved autophagic compartment and enhanced systemic elimination, offers lifelong protection from AD pathology by clearing toxic Aβ. Neuronal IGF-1R, and possibly other cell size-controlling pathways are promising targets for AD treatment. SIGNIFICANCE STATEMENT We found compelling evidence in vivo that Alzheimer's disease (AD) progression is significantly delayed when insulin-like growth factor (IGF) signaling is blocked in adult neurons. To show that, we built a novel mouse model, combining inducible neuron-specific IGF-1R knock-out with AD transgenics. Analysis of the experimental AD phenotype revealed less abundant amyloid-β (Aβ) peptides, fewer plaques, and diminished neuroinflammation in mutants with inactivated IGF signaling, together with clearly preserved behavioral and memory performances. We present for the first time evidence that IGF signaling has profound effects on neuronal proteostasis and maintenance of cell morphology in vivo. Our results indicate in a model highly pertinent to translational research that neuronal IGF resistance may represent a pathophysiologically relevant mechanism of the brain for preventing Aβ accumulation.
Collapse
|
68
|
Ayllon F, Kjærner-Semb E, Furmanek T, Wennevik V, Solberg MF, Dahle G, Taranger GL, Glover KA, Almén MS, Rubin CJ, Edvardsen RB, Wargelius A. The vgll3 Locus Controls Age at Maturity in Wild and Domesticated Atlantic Salmon (Salmo salar L.) Males. PLoS Genet 2015; 11:e1005628. [PMID: 26551894 PMCID: PMC4638356 DOI: 10.1371/journal.pgen.1005628] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/05/2015] [Indexed: 11/25/2022] Open
Abstract
Wild and domesticated Atlantic salmon males display large variation for sea age at sexual maturation, which varies between 1-5 years. Previous studies have uncovered a genetic predisposition for variation of age at maturity with moderate heritability, thus suggesting a polygenic or complex nature of this trait. The aim of this study was to identify associated genetic loci, genes and ultimately specific sequence variants conferring sea age at maturity in salmon. We performed a genome wide association study (GWAS) using a pool sequencing approach (20 individuals per river and phenotype) of male salmon returning to rivers as sexually mature either after one sea winter (2009) or three sea winters (2011) in six rivers in Norway. The study revealed one major selective sweep, which covered 76 significant SNPs in which 74 were found in a 370 kb region of chromosome 25. Genotyping other smolt year classes of wild and domesticated salmon confirmed this finding. Genotyping domesticated fish narrowed the haplotype region to four SNPs covering 2386 bp, containing the vgll3 gene, including two missense mutations explaining 33-36% phenotypic variation. A single locus was found to have a highly significant role in governing sea age at maturation in this species. The SNPs identified may be both used as markers to guide breeding for late maturity in salmon aquaculture and in monitoring programs of wild salmon. Interestingly, a SNP in proximity of the VGLL3 gene in humans (Homo sapiens), has previously been linked to age at puberty suggesting a conserved mechanism for timing of puberty in vertebrates.
Collapse
Affiliation(s)
| | - Erik Kjærner-Semb
- Institute of Marine Research, Bergen, Norway
- Department of Biology, University of Bergen, Bergen, Norway
| | | | | | | | - Geir Dahle
- Institute of Marine Research, Bergen, Norway
| | | | - Kevin A. Glover
- Institute of Marine Research, Bergen, Norway
- Department of Biology, University of Bergen, Bergen, Norway
| | - Markus Sällman Almén
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Carl J Rubin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
69
|
Clayton EL, Mizielinska S, Edgar JR, Nielsen TT, Marshall S, Norona FE, Robbins M, Damirji H, Holm IE, Johannsen P, Nielsen JE, Asante EA, Collinge J, Isaacs AM. Frontotemporal dementia caused by CHMP2B mutation is characterised by neuronal lysosomal storage pathology. Acta Neuropathol 2015; 130:511-23. [PMID: 26358247 PMCID: PMC4575387 DOI: 10.1007/s00401-015-1475-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 12/13/2022]
Abstract
Mutations in the charged multivesicular body protein 2B (CHMP2B) cause frontotemporal dementia (FTD). We report that mice which express FTD-causative mutant CHMP2B at physiological levels develop a novel lysosomal storage pathology characterised by large neuronal autofluorescent aggregates. The aggregates are an early and progressive pathology that occur at 3 months of age and increase in both size and number over time. These autofluorescent aggregates are not observed in mice expressing wild-type CHMP2B, or in non-transgenic controls, indicating that they are a specific pathology caused by mutant CHMP2B. Ultrastructural analysis and immuno- gold labelling confirmed that they are derived from the endolysosomal system. Consistent with these findings, CHMP2B mutation patient brains contain morphologically similar autofluorescent aggregates. These aggregates occur significantly more frequently in human CHMP2B mutation brain than in neurodegenerative disease or age-matched control brains. These data suggest that lysosomal storage pathology is the major neuronal pathology in FTD caused by CHMP2B mutation. Recent evidence suggests that two other genes associated with FTD, GRN and TMEM106B are important for lysosomal function. Our identification of lysosomal storage pathology in FTD caused by CHMP2B mutation now provides evidence that endolysosomal dysfunction is a major degenerative pathway in FTD.
Collapse
Affiliation(s)
- Emma L Clayton
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Sarah Mizielinska
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - James R Edgar
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Troels Tolstrup Nielsen
- Neurogenetics Research Laboratory, Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Marshall
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Frances E Norona
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Miranda Robbins
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Hana Damirji
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Ida E Holm
- Laboratory for Experimental Neuropathology, Department of Pathology, Randers Hospital, 8930, Randers NØ, Denmark
- Institute of Clinical Medicine, Aarhus University, 8000, Aarhus C, Denmark
| | - Peter Johannsen
- Memory Clinic, Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen E Nielsen
- Neurogenetics Research Laboratory, Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Memory Clinic, Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Section of Neurogenetics, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Emmanuel A Asante
- MRC Prion Unit, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - John Collinge
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- MRC Prion Unit, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK.
| |
Collapse
|
70
|
|
71
|
Fontana F, Siva K, Denti MA. A network of RNA and protein interactions in Fronto Temporal Dementia. Front Mol Neurosci 2015; 8:9. [PMID: 25852467 PMCID: PMC4365750 DOI: 10.3389/fnmol.2015.00009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/25/2015] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disorder characterized by degeneration of the fronto temporal lobes and abnormal protein inclusions. It exhibits a broad clinicopathological spectrum and has been linked to mutations in seven different genes. We will provide a picture, which connects the products of these genes, albeit diverse in nature and function, in a network. Despite the paucity of information available for some of these genes, we believe that RNA processing and post-transcriptional regulation of gene expression might constitute a common theme in the network. Recent studies have unraveled the role of mutations affecting the functions of RNA binding proteins and regulation of microRNAs. This review will combine all the recent findings on genes involved in the pathogenesis of FTD, highlighting the importance of a common network of interactions in order to study and decipher the heterogeneous clinical manifestations associated with FTD. This approach could be helpful for the research of potential therapeutic strategies.
Collapse
Affiliation(s)
- Francesca Fontana
- Laboratory of RNA Biology and Biotechnology, Centre for Integrative Biology, University of TrentoTrento, Italy
| | - Kavitha Siva
- Laboratory of RNA Biology and Biotechnology, Centre for Integrative Biology, University of TrentoTrento, Italy
| | - Michela A. Denti
- Laboratory of RNA Biology and Biotechnology, Centre for Integrative Biology, University of TrentoTrento, Italy
- CNR, Institute of NeurosciencePadua, Italy
| |
Collapse
|
72
|
Chassefeyre R, Martínez-Hernández J, Bertaso F, Bouquier N, Blot B, Laporte M, Fraboulet S, Couté Y, Devoy A, Isaacs AM, Pernet-Gallay K, Sadoul R, Fagni L, Goldberg Y. Regulation of postsynaptic function by the dementia-related ESCRT-III subunit CHMP2B. J Neurosci 2015; 35:3155-73. [PMID: 25698751 PMCID: PMC4331633 DOI: 10.1523/jneurosci.0586-14.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 12/23/2014] [Accepted: 01/06/2015] [Indexed: 12/14/2022] Open
Abstract
The charged multivesicular body proteins (Chmp1-7) are an evolutionarily conserved family of cytosolic proteins that transiently assembles into helical polymers that change the curvature of cellular membrane domains. Mutations in human CHMP2B cause frontotemporal dementia, suggesting that this protein may normally control some neuron-specific process. Here, we examined the function, localization, and interactions of neuronal Chmp2b. The protein was highly expressed in mouse brain and could be readily detected in neuronal dendrites and spines. Depletion of endogenous Chmp2b reduced dendritic branching of cultured hippocampal neurons, decreased excitatory synapse density in vitro and in vivo, and abolished activity-induced spine enlargement and synaptic potentiation. To understand the synaptic effects of Chmp2b, we determined its ultrastructural distribution by quantitative immuno-electron microscopy and its biochemical interactions by coimmunoprecipitation and mass spectrometry. In the hippocampus in situ, a subset of neuronal Chmp2b was shown to concentrate beneath the perisynaptic membrane of dendritic spines. In synaptoneurosome lysates, Chmp2b was stably bound to a large complex containing other members of the Chmp family, as well as postsynaptic scaffolds. The supramolecular Chmp assembly detected here corresponds to a stable form of the endosomal sorting complex required for transport-III (ESCRT-III), a ubiquitous cytoplasmic protein complex known to play a central role in remodeling of lipid membranes. We conclude that Chmp2b-containing ESCRT-III complexes are also present at dendritic spines, where they regulate synaptic plasticity. We propose that synaptic ESCRT-III filaments may function as a novel element of the submembrane cytoskeleton of spines.
Collapse
Affiliation(s)
- Romain Chassefeyre
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - José Martínez-Hernández
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Federica Bertaso
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34094 Montpellier, France, Universités de Montpellier 1 & 2, UMR-5203, F-34094 Montpellier, France, INSERM, Unité 661, F-34094 Montpellier, France
| | - Nathalie Bouquier
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34094 Montpellier, France, Universités de Montpellier 1 & 2, UMR-5203, F-34094 Montpellier, France, INSERM, Unité 661, F-34094 Montpellier, France
| | - Béatrice Blot
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Marine Laporte
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Sandrine Fraboulet
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Yohann Couté
- INSERM, Unité 1038, F-38054 Grenoble, France, Commissariat à l'Energie Atomique (CEA), Institut de Recherches en Technologies et Sciences pour le Vivant (iRTSV), Laboratoire de Biologie à Grande Echelle, F-38054 Grenoble, France
| | - Anny Devoy
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London WC1N 3BG, United Kingdom, and
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London WC1N 3BG, United Kingdom, and
| | - Karin Pernet-Gallay
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Rémy Sadoul
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France,
| | - Laurent Fagni
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34094 Montpellier, France, Universités de Montpellier 1 & 2, UMR-5203, F-34094 Montpellier, France, INSERM, Unité 661, F-34094 Montpellier, France
| | - Yves Goldberg
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France, CEA, iRTSV, Groupe Physiopathologie du Cytosquelette (GPC), F-38054 Grenoble, France
| |
Collapse
|
73
|
Dual Role of Autophagy in Neurodegenerative Diseases: The Case of Amyotrophic Lateral Sclerosis. CURRENT TOPICS IN NEUROTOXICITY 2015. [DOI: 10.1007/978-3-319-13939-5_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
74
|
|
75
|
Hardy J, Rogaeva E. Motor neuron disease and frontotemporal dementia: sometimes related, sometimes not. Exp Neurol 2014; 262 Pt B:75-83. [DOI: 10.1016/j.expneurol.2013.11.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 10/27/2013] [Accepted: 11/07/2013] [Indexed: 12/12/2022]
|
76
|
Gascon E, Lynch K, Ruan H, Almeida S, Verheyden J, Seeley WW, Dickson DW, Petrucelli L, Sun D, Jiao J, Zhou H, Jakovcevski M, Akbarian S, Yao WD, Gao FB. Alterations in microRNA-124 and AMPA receptors contribute to social behavioral deficits in frontotemporal dementia. Nat Med 2014; 20:1444-51. [PMID: 25401692 PMCID: PMC4257887 DOI: 10.1038/nm.3717] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/11/2014] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases, such as frontotemporal dementia (FTD), are often associated with behavioral deficits, but the underlying anatomical and molecular causes remain poorly understood. Here we show that forebrain-specific expression of FTD-associated mutant CHMP2B in mice causes several age-dependent neurodegenerative phenotypes, including social behavioral impairments. The social deficits were accompanied by a change in AMPA receptor (AMPAR) composition, leading to an imbalance between Ca(2+)-permeable and Ca(2+)-impermeable AMPARs. Expression of most AMPAR subunits was regulated by the brain-enriched microRNA miR-124, whose abundance was markedly decreased in the superficial layers of the cerebral cortex of mice expressing the mutant CHMP2B. We found similar changes in miR-124 and AMPAR levels in the frontal cortex and induced pluripotent stem cell-derived neurons from subjects with behavioral variant FTD. Moreover, ectopic miR-124 expression in the medial prefrontal cortex of mutant mice decreased AMPAR levels and partially rescued behavioral deficits. Knockdown of the AMPAR subunit Gria2 also alleviated social impairments. Our results identify a previously undescribed mechanism involving miR-124 and AMPARs in regulating social behavior in FTD and suggest a potential therapeutic avenue.
Collapse
Affiliation(s)
- Eduardo Gascon
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605 USA
| | - Kelleen Lynch
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605 USA
| | - Hongyu Ruan
- Division of Neurosciences, New England Primate Research Center, Harvard Medical School, Southborough, MA, 01772 USA
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605 USA
| | - Jamie Verheyden
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 USA
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA 94143, USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Danqiong Sun
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 USA
| | - Jian Jiao
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158 USA
| | - Hongru Zhou
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605 USA
| | - Mira Jakovcevski
- Brudnick Neuropsychiatric Research Institute, Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Schahram Akbarian
- Brudnick Neuropsychiatric Research Institute, Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Wei-Dong Yao
- Division of Neurosciences, New England Primate Research Center, Harvard Medical School, Southborough, MA, 01772 USA
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605 USA
| |
Collapse
|
77
|
RNA-Binding Proteins Associated Molecular Mechanisms of Motor Neuron Degeneration Pathogenesis. Mol Biotechnol 2014; 56:779-86. [DOI: 10.1007/s12033-014-9785-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
78
|
Frontotemporal Lobar Degeneration: Genetics and Clinical Phenotypes. NEURODEGENER DIS 2014. [DOI: 10.1007/978-1-4471-6380-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
79
|
Alzheimer’s Disease and Frontotemporal Lobar Degeneration: Mouse Models. NEURODEGENER DIS 2014. [DOI: 10.1007/978-1-4471-6380-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
80
|
Ling SC, Polymenidou M, Cleveland DW. Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron 2013; 79:416-38. [PMID: 23931993 DOI: 10.1016/j.neuron.2013.07.033] [Citation(s) in RCA: 1330] [Impact Index Per Article: 110.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2013] [Indexed: 12/12/2022]
Abstract
Breakthrough discoveries identifying common genetic causes for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) have transformed our view of these disorders. They share unexpectedly similar signatures, including dysregulation in common molecular players including TDP-43, FUS/TLS, ubiquilin-2, VCP, and expanded hexanucleotide repeats within the C9ORF72 gene. Dysfunction in RNA processing and protein homeostasis is an emerging theme. We present the case here that these two processes are intimately linked, with disease-initiated perturbation of either leading to further deviation of both protein and RNA homeostasis through a feedforward loop including cell-to-cell prion-like spread that may represent the mechanism for relentless disease progression.
Collapse
Affiliation(s)
- Shuo-Chien Ling
- Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, CA 92093-0670, USA
| | | | | |
Collapse
|
81
|
Cheruiyot A, Lee JA, Gao FB, Ahmad ST. Expression of mutant CHMP2B, an ESCRT-III component involved in frontotemporal dementia, causes eye deformities due to Notch misregulation in Drosophila. FASEB J 2013; 28:667-75. [PMID: 24158394 DOI: 10.1096/fj.13-234138] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Endosomal sorting complexes required for transport (ESCRTs) mediate sorting of ubiquitinated membrane proteins into multivesicular bodies en route to lysosomes for degradation. A mutation in CHMP2B (CHMP2B(Intron5), an ESCRT-III component) that is associated with a hereditary form of frontotemporal dementia (FTD3) disrupts the endosomal-lysosomal pathway and causes accumulation of autophagosomes and multilamellar structures. We previously demonstrated that expression of CHMP2B(Intron5) in the Drosophila eye using GMR-Gal4 causes misregulation of the Toll receptor pathway. Here, we show that ectopic expression of CHMP2B(Intron5) using eyeless-Gal4 (ey>CHMP2B(Intron5)), a driver with different spatiotemporal expression attributes than GMR-Gal4 in the Drosophila eye, causes eye deformities when compared to expression of wild-type CHMP2B (CHMP2B(WT)) and the Drosophila homologue of CHMP2B (CG4618). In addition, ey>CHMP2B(Intron5) flies showed defects in photoreceptor cell patterning and phototactic behavior. Furthermore, ey>CHMP2B(Intron5) flies showed accumulation of Notch in enlarged endosomes and up-regulation of Notch activity. Partial loss of Notch activity in ey>CHMP2B(Intron5) flies significantly rescued eye deformities, photoreceptor patterning defect, and phototactic behavior defect, indicating that these defects are primarily due to Notch misregulation. These results demonstrate that CHMP2B(Intron5) preferentially affects different receptor signaling pathways in a cellular and developmental context-dependent manner.
Collapse
Affiliation(s)
- Abigael Cheruiyot
- 2Department of Biology, Colby College, 5720 Mayflower Hill Dr., Waterville, ME 04901, USA.
| | | | | | | |
Collapse
|
82
|
Chen S, Sayana P, Zhang X, Le W. Genetics of amyotrophic lateral sclerosis: an update. Mol Neurodegener 2013; 8:28. [PMID: 23941283 PMCID: PMC3766231 DOI: 10.1186/1750-1326-8-28] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/29/2013] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder involving both upper motor neurons (UMN) and lower motor neurons (LMN). Enormous research has been done in the past few decades in unveiling the genetics of ALS, successfully identifying at least fifteen candidate genes associated with familial and sporadic ALS. Numerous studies attempting to define the pathogenesis of ALS have identified several plausible determinants and molecular pathways leading to motor neuron degeneration, which include oxidative stress, glutamate excitotoxicity, apoptosis, abnormal neurofilament function, protein misfolding and subsequent aggregation, impairment of RNA processing, defects in axonal transport, changes in endosomal trafficking, increased inflammation, and mitochondrial dysfunction. This review is to update the recent discoveries in genetics of ALS, which may provide insight information to help us better understanding of the disease neuropathogenesis.
Collapse
Affiliation(s)
- Sheng Chen
- Institute of Neurology, Jiao Tong University School of Medicine, 1201 Room, 11 Building, Ruijin Er Road, Shanghai 200025, China.
| | | | | | | |
Collapse
|
83
|
Roberson ED. Mouse models of frontotemporal dementia. Ann Neurol 2013; 72:837-49. [PMID: 23280835 DOI: 10.1002/ana.23722] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 07/05/2012] [Accepted: 07/30/2012] [Indexed: 12/14/2022]
Abstract
The pace of discovery in frontotemporal dementia (FTD) has accelerated dramatically with the discovery of new genetic causes and pathological substrates of the disease. MAPT/tau, GRN/progranulin, and C9ORF72 have emerged as common FTD genes, and TARDBP/TDP-43, VCP, FUS, and CHMP2B have been identified as less common genetic causes. TDP-43 and FUS have joined tau as common neuropathological substrates of the disease. Mouse models provide an important tool for understanding the role of these molecules in FTD pathogenesis. Here, we review recent progress with mouse models based on tau, TDP-43, progranulin, VCP, and CHMP2B. We also consider future prospects for FTD models, including developing new models to address unanswered questions. There are also opportunities for capitalizing on conservation of the salience network, which is selectively vulnerable in FTD, and the availability of FTD-related behavioral paradigms to analyze mouse models of the disease.
Collapse
Affiliation(s)
- Erik D Roberson
- Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama, Birmingham, AL 35294, USA.
| |
Collapse
|
84
|
Abstract
Frontotemporal dementia (FTD) is the second most common type of presenile dementia and is the most common form of dementia with the onset before 60 years of age. Its typical symptoms include behavioral disorders, affective symptoms, and language disorders. The FTD is a genetically and pathologically heterogeneous degenerative disorder. Animal models have provided more insights into the pathogenic mechanisms. There are currently no medications that are specifically approved for the treatment of FTD by the Food and Drug Administration. In this article, we review the recent advances in the molecular pathogenesis, pathology, animal models, and therapy for FTD. Better understanding of the pathogenesis and the use of animal models will help develop novel therapeutic strategies and provide new targets for FTD treatment.
Collapse
Affiliation(s)
- Xinling Wang
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Medical Neurobiology of Chinese Ministry of Health, Hangzhou, China
| | - Yuedi Shen
- Center for Cognition and Brain Disorders & The Affiliated Hospital, Hangzhou Normal University School, Hangzhou, China
| | - Wei Chen
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Medical Neurobiology of Chinese Ministry of Health, Hangzhou, China
| |
Collapse
|
85
|
Nielsen TT, Mizielinska S, Hasholt L, Isaacs AM, Nielsen JE. Reversal of pathology in CHMP2B-mediated frontotemporal dementia patient cells using RNA interference. J Gene Med 2013; 14:521-9. [PMID: 22786763 DOI: 10.1002/jgm.2649] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Frontotemporal dementia is the second most common form of young-onset dementia after Alzheimer's disease, and several genetic forms of frontotemporal dementia are known. A rare genetic variant is caused by a point mutation in the CHMP2B gene. CHMP2B is a component of the ESCRT-III complex, which is involved in endosomal trafficking of proteins targeted for degradation in lysosomes. Mutations in CHMP2B result in abnormal endosomal structures in patient fibroblasts and patient brains, probably through a gain-of-function mechanism, suggesting that the endosomal pathway plays a central role in the pathogenesis of the disease. METHODS In the present study, we used lentiviral vectors to efficiently knockdown CHMP2B by delivering microRNA embedded small hairpin RNAs. RESULTS We show that CHMP2B can be efficiently knocked down in patient fibroblasts using an RNA interference approach and that the knockdown causes reversal of the abnormal endosomal phenotype observed in patient fibroblasts. CONCLUSIONS This is the first description of a treatment that reverses the cellular pathology caused by mutant CHMP2B and suggests that RNA interference might be a feasible therapeutic strategy. Furthermore, it provides the first proof of a direct link between the disease-causing mutation and the cellular phenotype in cells originating from CHMP2B mutation patients.
Collapse
Affiliation(s)
- Troels Tolstrup Nielsen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | | | | | | | | | | |
Collapse
|
86
|
Xu J, Nonogaki M, Madhira R, Ma HY, Hermanson O, Kioussi C, Gross MK. Population-specific regulation of Chmp2b by Lbx1 during onset of synaptogenesis in lateral association interneurons. PLoS One 2012; 7:e48573. [PMID: 23284619 PMCID: PMC3528757 DOI: 10.1371/journal.pone.0048573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 09/27/2012] [Indexed: 12/12/2022] Open
Abstract
Chmp2b is closely related to Vps2, a key component of the yeast protein complex that creates the intralumenal vesicles of multivesicular bodies. Dominant negative mutations in Chmp2b cause autophagosome accumulation and neurodegenerative disease. Loss of Chmp2b causes failure of dendritic spine maturation in cultured neurons. The homeobox gene Lbx1 plays an essential role in specifying postmitotic dorsal interneuron populations during late pattern formation in the neural tube. We have discovered that Chmp2b is one of the most highly regulated cell-autonomous targets of Lbx1 in the embryonic mouse neural tube. Chmp2b was expressed and depended on Lbx1 in only two of the five nascent, Lbx1-expressing, postmitotic, dorsal interneuron populations. It was also expressed in neural tube cell populations that lacked Lbx1 protein. The observed population-specific expression of Chmp2b indicated that only certain population-specific combinations of sequence specific transcription factors allow Chmp2b expression. The cell populations that expressed Chmp2b corresponded, in time and location, to neurons that make the first synapses of the spinal cord. Chmp2b protein was transported into neurites within the motor- and association-neuropils, where the first synapses are known to form between E11.5 and E12.5 in mouse neural tubes. Selective, developmentally-specified gene expression of Chmp2b may therefore be used to endow particular neuronal populations with the ability to mature dendritic spines. Such a mechanism could explain how mammalian embryos reproducibly establish the disynaptic cutaneous reflex only between particular cell populations.
Collapse
Affiliation(s)
- Jun Xu
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Mariko Nonogaki
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Ravi Madhira
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Hsiao-Yen Ma
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Michael K. Gross
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, Oregon, United States of America
- * E-mail:
| |
Collapse
|
87
|
Endosomal sorting related protein CHMP2B is localized in Lewy bodies and glial cytoplasmic inclusions in α-synucleinopathy. Neurosci Lett 2012; 527:16-21. [DOI: 10.1016/j.neulet.2012.08.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 08/10/2012] [Accepted: 08/17/2012] [Indexed: 12/23/2022]
|
88
|
Han JH, Ryu HH, Jun MH, Jang DJ, Lee JA. The functional analysis of the CHMP2B missense mutation associated with neurodegenerative diseases in the endo-lysosomal pathway. Biochem Biophys Res Commun 2012; 421:544-9. [PMID: 22521643 DOI: 10.1016/j.bbrc.2012.04.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 04/07/2012] [Indexed: 12/12/2022]
Abstract
Endosomal sorting complexes required for transport (ESCRTs) regulate a key sorting step of protein trafficking between endosomal compartments in lysosomal degradation. Interestingly, mutations in charged multivesicular body protein 2B (CHMP2B), which is a core subunit of ESCRT-III, have been identified in some neurodegenerative diseases. However, the cellular pathogenesis resulting from CHMP2B missense mutations is unclear. Furthermore, little is known about their functional analysis in post-mitotic neurons. In order to examine their cellular pathogenesis, we analyzed their effects in the endo-lysosomal pathway in post-mitotic neurons. Interestingly, of the missense mutant proteins, CHMP2B(T104N) mostly accumulated in the Rab5- and Rab7-positive endosomes and caused delayed degradation of EGFR as compared to CHMP2B(WT). Furthermore, CHMP2B(T104N) showed less association with Vps4 ATPase and was avidly associated with Snf7-2, a core component of ESCRT-III, suggesting that it may cause defects in the process of dissociation from ESCRT. Of the missense variants, CHMP2B(T104N) caused prominent accumulation of autophagosomes. However, neuronal cell survival was not dramatically affected by expression of CHMP2B(T104N). These findings suggested that, from among the various missense mutants, CHMP2B(T104N) was associated with relatively mild cellular pathogenesis in post-mitotic neurons. This study provided a better understanding of the cellular pathogenesis of neurodegenerative diseases associated with various missense mutations of CHMP2B as well as endocytic defects.
Collapse
Affiliation(s)
- Jeong-Ho Han
- Department of Biotechnology, College of Life Science and Nano Technology, Hannam University, 461-6, Jeonmin-dong, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | | | | | | | | |
Collapse
|
89
|
Abstract
Gene products such as organelles, proteins and RNAs are actively transported to synaptic terminals for the remodeling of pre-existing neuronal connections and formation of new ones. Proteins described as molecular motors mediate this transport and utilize specialized cytoskeletal proteins that function as molecular tracks for the motor based transport of cargos. Molecular motors such as kinesins and dynein's move along microtubule tracks formed by tubulins whereas myosin motors utilize tracks formed by actin. Deficits in active transport of gene products have been implicated in a number of neurological disorders. We describe such disorders collectively as "transportopathies". Here we review current knowledge of critical components of active transport and their relevance to neurodegenerative diseases.
Collapse
|