1
|
Xie L, Zhu Y, Hurtle BT, Wright M, Robinson JL, Mauna JC, Brown EE, Ngo M, Bergmann CA, Xu J, Merjane J, Gleixner AM, Grigorean G, Liu F, Rossoll W, Lee EB, Kiskinis E, Chikina M, Donnelly CJ. Context-dependent Interactors Regulate TDP-43 Dysfunction in ALS/FTLD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.646890. [PMID: 40291645 PMCID: PMC12026901 DOI: 10.1101/2025.04.07.646890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
TDP-43 mislocalization, aggregation, and loss of splicing function are neuropathological hallmarks in over 97% of Amyotrophic Lateral Sclerosis (ALS), 45% of Frontotemporal Lobar Degeneration (FTLD), and 60% of Alzheimer's Disease, which has been reclassified as LATE-NC. However, the mechanisms underlying TDP-43 dysfunction remain elusive. Here, we utilize APEX2-driven proximity labeling and mass spectrometry to characterize the context-dependent TDP-43 interactome in conditions of cytoplasmic mislocalization, impaired RNA-binding contributing to aggregation, and oxidative stress. We describe context-dependent interactors, including disrupted interactions with splicing-related proteins and altered biomolecular condensate (BMC) associations. By integrating ALS and FTLD snRNA-seq data, we uncover disease-relevant molecular alterations and validate our dataset through a functional screen that identifies key TDP- 43 regulators. We demonstrate that disrupting nuclear speckle integrity, particularly through the downregulation of the splicing factor SRRM2, promotes TDP-43 mislocalization and loss of function. Additionally, we identify NUFIP2 as an interactor associated with mislocalization that sequesters TDP-43 into cytoplasmic aggregates and co-localizes with TDP-43 pathology in patient tissue. We also highlight HNRNPC as a potent TDP-43 splicing regulator, where precise modulation of TDP-43 or HNRNPC can rescue cryptic exon splicing. These findings provide mechanistic insights and potential therapeutic targets for TDP-43 dysfunction.
Collapse
|
2
|
Biswas DD, Sethi R, Woldeyohannes Y, Scarrow ER, El Haddad L, Lee J, ElMallah MK. Respiratory pathology in the TDP-43 transgenic mouse model of amyotrophic lateral sclerosis. Front Physiol 2024; 15:1430875. [PMID: 39403566 PMCID: PMC11471906 DOI: 10.3389/fphys.2024.1430875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/29/2024] [Indexed: 03/28/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that results in death within 2-5 years of diagnosis. Respiratory failure is the most common cause of death in ALS. Mutations in the transactive response DNA binding protein 43 (TDP-43) encoded by the TARDBP gene are associated with abnormal cellular aggregates in neurons of patients with both familial and sporadic ALS. The role of these abnormal aggregates on breathing is unclear. Since respiratory failure is a major cause of death in ALS, we sought to determine the role of TDP-43 mutations on the respiratory motor unit in the Prp-hTDP-43A315T mouse model - a model that expresses human TDP-43 containing the A315T mutation. We assessed breathing using whole-body plethysmography, and investigated neuropathology in hypoglossal and phrenic respiratory motor units. Postmortem studies included quantification of hypoglossal and putative phrenic motor neurons, activated microglia and astrocytes in respiratory control centers, and assessment of hypoglossal and phrenic nerves of TDP43A315T mice. The male TDP43A315T mice display an early onset of rapid progression of disease, and premature death (less than 15 weeks) compared to control mice and compared to female TDP43A315T mice who die between 20 and 35 weeks of age. The TDP43A315T mice have progressive and profound breathing deficits at baseline and during a respiratory challenge. Histologically, hypoglossal and putative phrenic motor neurons of TDP43A315T mice are decreased and have increased microglial and astrocyte activation, indicating pronounced neurodegeneration and neuroinflammation. Further, there is axonopathy and demyelination in the hypoglossal and phrenic nerve of TDP43A315T mice. Thus, the TDP-43A315T mice have significant respiratory pathology and neuropathology, which makes them a useful translatable model for the study of novel therapies on breathing in ALS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mai K. ElMallah
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
3
|
Carmen-Orozco RP, Tsao W, Ye Y, Sinha IR, Chang K, Trinh VT, Chung W, Bowden K, Troncoso JC, Blackshaw S, Hayes LR, Sun S, Wong PC, Ling JP. Elevated nuclear TDP-43 induces constitutive exon skipping. Mol Neurodegener 2024; 19:45. [PMID: 38853250 PMCID: PMC11163724 DOI: 10.1186/s13024-024-00732-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 05/20/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Cytoplasmic inclusions and loss of nuclear TDP-43 are key pathological features found in several neurodegenerative disorders, suggesting both gain- and loss-of-function mechanisms of disease. To study gain-of-function, TDP-43 overexpression has been used to generate in vitro and in vivo model systems. METHODS We analyzed RNA-seq datasets from mouse and human neurons overexpressing TDP-43 to explore species specific splicing patterns. We explored the dynamics between TDP-43 levels and exon repression in vitro. Furthermore we analyzed human brain samples and publicly available RNA datasets to explore the relationship between exon repression and disease. RESULTS Our study shows that excessive levels of nuclear TDP-43 protein lead to constitutive exon skipping that is largely species-specific. Furthermore, while aberrant exon skipping is detected in some human brains, it is not correlated with disease, unlike the incorporation of cryptic exons that occurs after loss of TDP-43. CONCLUSIONS Our findings emphasize the need for caution in interpreting TDP-43 overexpression data and stress the importance of controlling for exon skipping when generating models of TDP-43 proteinopathy.
Collapse
Affiliation(s)
- Rogger P Carmen-Orozco
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - William Tsao
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Yingzhi Ye
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Irika R Sinha
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Koping Chang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Vickie T Trinh
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - William Chung
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Kyra Bowden
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Juan C Troncoso
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Lindsey R Hayes
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Shuying Sun
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Philip C Wong
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Jonathan P Ling
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
4
|
Cantara S, Simoncelli G, Ricci C. Antisense Oligonucleotides (ASOs) in Motor Neuron Diseases: A Road to Cure in Light and Shade. Int J Mol Sci 2024; 25:4809. [PMID: 38732027 PMCID: PMC11083842 DOI: 10.3390/ijms25094809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Antisense oligonucleotides (ASOs) are short oligodeoxynucleotides designed to bind to specific regions of target mRNA. ASOs can modulate pre-mRNA splicing, increase levels of functional proteins, and decrease levels of toxic proteins. ASOs are being developed for the treatment of motor neuron diseases (MNDs), including spinal muscular atrophy (SMA), amyotrophic lateral sclerosis (ALS) and spinal and bulbar muscular atrophy (SBMA). The biggest success has been the ASO known as nusinersen, the first effective therapy for SMA, able to improve symptoms and slow disease progression. Another success is tofersen, an ASO designed to treat ALS patients with SOD1 gene mutations. Both ASOs have been approved by the FDA and EMA. On the other hand, ASO treatment in ALS patients with the C9orf72 gene mutation did not show any improvement in disease progression. The aim of this review is to provide an up-to-date overview of ASO research in MNDs, from preclinical studies to clinical trials and, where available, regulatory approval. We highlight the successes and failures, underline the strengths and limitations of the current ASO research, and suggest possible approaches that could lead to more effective treatments.
Collapse
Affiliation(s)
- Silvia Cantara
- Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy;
| | - Giorgia Simoncelli
- Unit of Neurology and Clinical Neurophysiology, Department of Neurological and Motor Sciences, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy;
| | - Claudia Ricci
- Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy;
| |
Collapse
|
5
|
Van Daele SH, Masrori P, Van Damme P, Van Den Bosch L. The sense of antisense therapies in ALS. Trends Mol Med 2024; 30:252-262. [PMID: 38216448 DOI: 10.1016/j.molmed.2023.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024]
Abstract
Treatment of patients with amyotrophic lateral sclerosis (ALS) has entered a new era now that encouraging results about antisense oligonucleotides (ASOs) are becoming available and a first ASO therapy for ALS has been approved by the FDA. Moreover, there is hope not only that ALS can be stopped but also that symptoms can be reversed. Until now, degrading ASOs seemed to be successful mostly for rarer forms of familial ALS. However, the first attempts to correct mis-splicing events in sporadic ALS are underway, as well as a clinical trial examining interference with a genetic modifier. In this review, we discuss the current status of using ASOs in ALS and the possibilities and pitfalls of this therapeutic strategy.
Collapse
Affiliation(s)
- Sien H Van Daele
- KU Leuven - University of Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), Leuven, Belgium; Laboratory of Neurobiology, VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Pegah Masrori
- KU Leuven - University of Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), Leuven, Belgium; Laboratory of Neurobiology, VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Philip Van Damme
- KU Leuven - University of Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), Leuven, Belgium; Laboratory of Neurobiology, VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurology, University Hospitals Leuven, Leuven, Belgium.
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), Leuven, Belgium; Laboratory of Neurobiology, VIB Center for Brain & Disease Research, Leuven, Belgium.
| |
Collapse
|
6
|
Markovinovic A, Martín-Guerrero SM, Mórotz GM, Salam S, Gomez-Suaga P, Paillusson S, Greig J, Lee Y, Mitchell JC, Noble W, Miller CCJ. Stimulating VAPB-PTPIP51 ER-mitochondria tethering corrects FTD/ALS mutant TDP43 linked Ca 2+ and synaptic defects. Acta Neuropathol Commun 2024; 12:32. [PMID: 38395965 PMCID: PMC10885568 DOI: 10.1186/s40478-024-01742-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are clinically linked major neurodegenerative diseases. Notably, TAR DNA-binding protein-43 (TDP43) accumulations are hallmark pathologies of FTD/ALS and mutations in the gene encoding TDP43 cause familial FTD/ALS. There are no cures for FTD/ALS. FTD/ALS display damage to a broad range of physiological functions, many of which are regulated by signaling between the endoplasmic reticulum (ER) and mitochondria. This signaling is mediated by the VAPB-PTPIP51 tethering proteins that serve to recruit regions of ER to the mitochondrial surface so as to facilitate inter-organelle communications. Several studies have now shown that disrupted ER-mitochondria signaling including breaking of the VAPB-PTPIP51 tethers are features of FTD/ALS and that for TDP43 and other familial genetic FTD/ALS insults, this involves activation of glycogen kinase-3β (GSK3β). Such findings have prompted suggestions that correcting damage to ER-mitochondria signaling and the VAPB-PTPIP51 interaction may be broadly therapeutic. Here we provide evidence to support this notion. We show that overexpression of VAPB or PTPIP51 to enhance ER-mitochondria signaling corrects mutant TDP43 induced damage to inositol 1,4,5-trisphosphate (IP3) receptor delivery of Ca2+ to mitochondria which is a primary function of the VAPB-PTPIP51 tethers, and to synaptic function. Moreover, we show that ursodeoxycholic acid (UDCA), an FDA approved drug linked to FTD/ALS and other neurodegenerative diseases therapy and whose precise therapeutic target is unclear, corrects TDP43 linked damage to the VAPB-PTPIP51 interaction. We also show that this effect involves inhibition of TDP43 mediated activation of GSK3β. Thus, correcting damage to the VAPB-PTPIP51 tethers may have therapeutic value for FTD/ALS and other age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Andrea Markovinovic
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK.
| | - Sandra M Martín-Guerrero
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK
| | - Gábor M Mórotz
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK
| | - Shaakir Salam
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK
| | - Patricia Gomez-Suaga
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK
| | - Sebastien Paillusson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK
| | - Jenny Greig
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK
| | - Younbok Lee
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK
| | - Jacqueline C Mitchell
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK
| | - Christopher C J Miller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9RX, London, UK.
| |
Collapse
|
7
|
Ovsepian SV, O'Leary VB, Martinez S. Selective vulnerability of motor neuron types and functional groups to degeneration in amyotrophic lateral sclerosis: review of the neurobiological mechanisms and functional correlates. Brain Struct Funct 2024; 229:1-14. [PMID: 37999738 PMCID: PMC10827929 DOI: 10.1007/s00429-023-02728-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative condition characterised by a progressive loss of motor neurons controlling voluntary muscle activity. The disease manifests through a variety of motor dysfunctions related to the extent of damage and loss of neurons at different anatomical locations. Despite extensive research, it remains unclear why some motor neurons are especially susceptible to the disease, while others are affected less or even spared. In this article, we review the neurobiological mechanisms, neurochemical profiles, and morpho-functional characteristics of various motor neuron groups and types of motor units implicated in their differential exposure to degeneration. We discuss specific cell-autonomous (intrinsic) and extrinsic factors influencing the vulnerability gradient of motor units and motor neuron types to ALS, with their impact on disease manifestation, course, and prognosis, as revealed in preclinical and clinical studies. We consider the outstanding challenges and emerging opportunities for interpreting the phenotypic and mechanistic variability of the disease to identify targets for clinical interventions.
Collapse
Affiliation(s)
- Saak V Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, UK.
| | - Valerie B O'Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruská 87, 10000, Prague, Czech Republic
| | - Salvador Martinez
- Instituto de Neurociencias UMH-CSIC, Avda. Ramon y Cajal, 03550, San Juan de Alicante, Spain.
- Center of Biomedical Network Research on Mental Health (CIBERSAM), ISCIII, Madrid, Spain.
| |
Collapse
|
8
|
Ke H, Liu K, Jiao B, Zhao L. Implications of TDP-43 in non-neuronal systems. Cell Commun Signal 2023; 21:338. [PMID: 37996849 PMCID: PMC10666381 DOI: 10.1186/s12964-023-01336-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/26/2023] [Indexed: 11/25/2023] Open
Abstract
TAR DNA-binding protein 43 (TDP-43) is a versatile RNA/DNA-binding protein with multifaceted processes. While TDP-43 has been extensively studied in the context of degenerative diseases, recent evidence has also highlighted its crucial involvement in diverse life processes beyond neurodegeneration. Here, we mainly reviewed the function of TDP-43 in non-neurodegenerative physiological and pathological processes, including spermatogenesis, embryonic development, mammary gland development, tumor formation, and viral infection, highlighting its importance as a key regulatory factor for the maintenance of normal functions throughout life. TDP-43 exhibits diverse and sometimes opposite functionality across different cell types through various mechanisms, and its roles can shift at distinct stages within the same biological system. Consequently, TDP-43 operates in both a context-dependent and a stage-specific manner in response to a variety of internal and external stimuli. Video Abstract.
Collapse
Affiliation(s)
- Hao Ke
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China
| | - Kang Liu
- Ganzhou People's Hospital, Ganzhou, 341000, China
| | - Baowei Jiao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650201, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Limin Zhao
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China.
| |
Collapse
|
9
|
Sammeta SS, Banarase TA, Rahangdale SR, Wankhede NL, Aglawe MM, Taksande BG, Mangrulkar SV, Upaganlawar AB, Koppula S, Kopalli SR, Umekar MJ, Kale MB. Molecular understanding of ER-MT communication dysfunction during neurodegeneration. Mitochondrion 2023; 72:59-71. [PMID: 37495165 DOI: 10.1016/j.mito.2023.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
Biological researchers are seeing organelles in a new light. These cellular entities have been believed to be singular and distinctive structures that performed specialized purposes for a very long time. But in recentpast years, scientists have learned that organelles become dynamic and make physical contact. Additionally, Biological processes are regulated by organelles interactions and its alteration play an important role in cell malfunctioning and several pathologies, including neurodegenerative diseases. Mitochondrial-ER contact sites (MERCS) have received considerable attention in the domain of cell homeostasis and dysfunction, specifically in the area of neurodegeneration. This is largely due to the significant role of this subcellular compartment in a diverse array of vital cellular functions, including Ca2+ homeostasis, transport, bioenergetics and turnover, mitochondrial dynamics, apoptotic signaling, ER stress, and inflammation. A significant number of disease-associated proteins were found to physically interact with the ER-Mitochondria (ER-MT) interface, causing structural and/or functional alterations in this compartment. In this review, we summarize current knowledge about the structure and functions of the ER-MT contact sites, as well as the possible repercussions of their alteration in notable neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and fronto-temporal dementia. The constraints and complexities in defining the nature and origin of the highlighted defects in ER-MT communication, as well as their concise contribution to the neurodegenerative process, are illustrated in particular. The possibility of using MERCS as a potential drug target to prevent neuronal damage and ultimately neurodegeneration is the topic of our final discussion.
Collapse
Affiliation(s)
- Shivkumar S Sammeta
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Trupti A Banarase
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip R Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Manish M Aglawe
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Shubhada V Mangrulkar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| |
Collapse
|
10
|
Genç B, Nho B, Seung H, Helmold B, Park H, Gözütok Ö, Kim S, Park J, Ye S, Lee H, Lee N, Yu SS, Kim S, Lee J, Özdinler H. Novel rAAV vector mediated intrathecal HGF delivery has an impact on neuroimmune modulation in the ALS motor cortex with TDP-43 pathology. Gene Ther 2023; 30:560-574. [PMID: 36823441 DOI: 10.1038/s41434-023-00383-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/21/2022] [Accepted: 01/23/2023] [Indexed: 02/25/2023]
Abstract
Recombinant adeno-associated virus (rAAV)-based gene therapies offer an immense opportunity for rare diseases, such as amyotrophic lateral sclerosis (ALS), which is defined by the loss of the upper and the lower motor neurons. Here, we describe generation, characterization, and utilization of a novel vector system, which enables expression of the active form of hepatocyte growth factor (HGF) under EF-1α promoter with bovine growth hormone (bGH) poly(A) sequence and is effective with intrathecal injections. HGF's role in promoting motor neuron survival had been vastly reported. Therefore, we investigated whether intrathecal delivery of HGF would have an impact on one of the most common pathologies of ALS: the TDP-43 pathology. Increased astrogliosis, microgliosis and progressive upper motor neuron loss are important consequences of ALS in the motor cortex with TDP-43 pathology. We find that cortex can be modulated via intrathecal injection, and that expression of HGF reduces astrogliosis, microgliosis in the motor cortex, and help restore ongoing UMN degeneration. Our findings not only introduce a novel viral vector for the treatment of ALS, but also demonstrate modulation of motor cortex by intrathecal viral delivery, and that HGF treatment is effective in reducing astrogliosis and microgliosis in the motor cortex of ALS with TDP-43 pathology.
Collapse
Affiliation(s)
- Barış Genç
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Boram Nho
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea
| | - Hana Seung
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea
| | - Benjamin Helmold
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Huiwon Park
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea
| | - Öge Gözütok
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Seunghyun Kim
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea
| | - Jinil Park
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea
| | - Sanghyun Ye
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea
| | - Haneul Lee
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea
| | - Nayeon Lee
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Seung-Shin Yu
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea
| | - Sunyoung Kim
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea
| | - Junghun Lee
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
- Helixmith Co., Ltd., R&D Center, 21, Magokjungang 8-ro 7-gil, Gangseo-gu, Seoul, 07794, Republic of Korea.
| | - Hande Özdinler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Ave, Chicago, IL, 60611, USA.
- Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, Evanston, IL, 60208, USA.
| |
Collapse
|
11
|
Carmen-Orozco RP, Tsao W, Ye Y, Sinha IR, Chang K, Trinh V, Chung W, Bowden K, Troncoso JC, Blackshaw S, Hayes LR, Sun S, Wong PC, Ling JP. Elevated nuclear TDP-43 induces constitutive exon skipping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.11.540291. [PMID: 37215013 PMCID: PMC10197708 DOI: 10.1101/2023.05.11.540291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Cytoplasmic inclusions and loss of nuclear TDP-43 are key pathological features found in several neurodegenerative disorders, suggesting both gain- and loss-of-function mechanisms of disease. To study gain-of-function, TDP-43 overexpression has been used to generate in vitro and in vivo model systems. Our study shows that excessive levels of nuclear TDP-43 protein lead to constitutive exon skipping that is largely species-specific. Furthermore, while aberrant exon skipping is detected in some human brains, it is not correlated with disease, unlike the incorporation of cryptic exons that occurs after loss of TDP-43. Our findings emphasize the need for caution in interpreting TDP-43 overexpression data, and stress the importance of controlling for exon skipping when generating models of TDP-43 proteinopathy. Understanding the subtle aspects of TDP-43 toxicity within different subcellular locations is essential for the development of therapies targeting neurodegenerative disease.
Collapse
Affiliation(s)
- Rogger P Carmen-Orozco
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Neuroscience Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - William Tsao
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Neuroscience Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Yingzhi Ye
- Department of Physiology Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Irika R Sinha
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Neuroscience Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Koping Chang
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Vickie Trinh
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Neuroscience Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - William Chung
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Kyra Bowden
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Juan C Troncoso
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Seth Blackshaw
- Department of Neuroscience Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Ophthalmology Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Neurology Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Lindsey R Hayes
- Department of Neurology Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Shuying Sun
- Department of Physiology Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Philip C Wong
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
- Department of Neuroscience Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Jonathan P Ling
- Department of Pathology Johns Hopkins School of Medicine, Baltimore, MD 21205
| |
Collapse
|
12
|
Günther R. [Gene Therapies in Motor Neuron Diseases ALS and SMA]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2023; 91:153-163. [PMID: 36822211 DOI: 10.1055/a-2002-5215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
In the past, the diagnosis of motor neuron diseases such as amyotrophic lateral sclerosis (ALS) and 5q-associated spinal muscular atrophy (SMA) meant powerlessness in the face of seemingly untreatable diseases with severe motor-functional limitations and sometimes fatal courses. Recent advances in an understanding of the genetic causalities of these diseases, combined with success in the development of targeted gene therapy strategies, spell hope for effective, innovative therapeutic approaches, pioneering the ability to treat neurodegenerative diseases. While gene therapies have been approved for SMA since a few years, gene therapy research in ALS is still in clinical trials with encouraging results. This article provides an overview of the genetic background of ALS and SMA known to date and gene therapy approaches to them with a focus on therapy candidates that are in clinical trials or have already gained market approval.
Collapse
Affiliation(s)
- René Günther
- Klinik und Poliklinik für Neurologie, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
13
|
Jiang L, Zhang T, Lu K, Qi S. The progress in C9orf72 research: ALS/FTD pathogenesis, functions and structure. Small GTPases 2022; 13:56-76. [PMID: 33663328 PMCID: PMC9707547 DOI: 10.1080/21541248.2021.1892443] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The hexanucleotide repeat (GGGGCC) expansion in C9orf72 is accounted for a large proportion of the genetic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The hypotheses of how the massive G4C2 repeats in C9orf72 destroy the neurons and lead to ALS/FTD are raised and improving. As a multirole player, C9orf72 exerts critical roles in many cellular processes, including autophagy, membrane trafficking, immune response, and so on. Notably, the partners of C9orf72, through which C9orf72 participates in the cell activities, have been identified. Notably, the structures of the C9orf72-SMCR8-WDR41 complex shed light on its activity as GTPase activating proteins (GAP). In this manuscript, we reviewed the latest research progress in the C9orf72-mediated ALS/FTD, the physiological functions of C9orf72, and the putative function models of C9orf72/C9orf72-containing complex.
Collapse
Affiliation(s)
- Lan Jiang
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China
| | - Tizhong Zhang
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China
| | - Kefeng Lu
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China
| | - Shiqian Qi
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China,CONTACT Shiqian Qi Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, Chengdu, China.
These authors contributed equally to this work.
| |
Collapse
|
14
|
Genetic architecture of motor neuron diseases. J Neurol Sci 2021; 434:120099. [PMID: 34965490 DOI: 10.1016/j.jns.2021.120099] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/26/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022]
Abstract
Motor neuron diseases (MNDs) are rare and frequently fatal neurological disorders in which motor neurons within the brainstem and spinal cord regions slowly die. MNDs are primarily caused by genetic mutations, and > 100 different mutant genes in humans have been discovered thus far. Given the fact that many more MND-related genes have yet to be discovered, the growing body of genetic evidence has offered new insights into the diverse cellular and molecular mechanisms involved in the aetiology and pathogenesis of MNDs. This search may aid in the selection of potential candidate genes for future investigation and, eventually, may open the door to novel interventions to slow down disease progression. In this review paper, we have summarized detailed existing research findings of different MNDs, such as amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), spinal bulbar muscle atrophy (SBMA) and hereditary spastic paraplegia (HSP) in relation to their complex genetic architecture.
Collapse
|
15
|
Phenotypic diversity in ALS and the role of poly-conformational protein misfolding. Acta Neuropathol 2021; 142:41-55. [PMID: 32930869 DOI: 10.1007/s00401-020-02222-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/30/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022]
Abstract
In many types of familial amyotrophic lateral sclerosis (fALS), mutations cause proteins to gain toxic properties that mediate neurodegenerative processes. It is becoming increasingly clear that the proteins involved in ALS, and those responsible for a host of other neurodegenerative diseases, share many characteristics with a growing number of prion diseases. ALS is a heterogenous disease in which the majority of cases are sporadic in their etiology. Studies investigating the inherited forms of the disease are now beginning to provide evidence that some of this heterogeneity may be due to the existence of distinct conformations that ALS-linked proteins can adopt to produce the equivalent of prion strains. In this review, we discuss the in vitro and in vivo evidence that has been generated to better understand the characteristics of these proteins and how their tertiary structure may impact the disease phenotype.
Collapse
|
16
|
Baralle M, Romano M. Characterization of the human TARDBP gene promoter. Sci Rep 2021; 11:10438. [PMID: 34002018 PMCID: PMC8129075 DOI: 10.1038/s41598-021-89973-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/04/2021] [Indexed: 02/03/2023] Open
Abstract
The expression of TDP-43, the main component of neuronal intracellular inclusions across a broad spectrum of ALS and FTD disorders, is developmentally regulated and studies in vivo have shown that TDP-43 overexpression can be toxic, even before observation of pathological aggregates. Starting from these observations, the regulation of its expression at transcriptional level might represent a further key element for the pathogenesis of neurodegenerative diseases. Therefore, we have characterized the human TARDBP promoter, in order to study the transcriptional mechanisms of expression. Mapping of cis-acting elements by luciferase assays in different cell outlined that the activity of the promoter seems to be higher in SH-SY5Y, Neuro2A, and HeLa than in HEK293. In addition, we tested effects of two SNPs found in the promoter region of ALS patients and observed no significant effect on transcription levels in all tested cell lines. Lastly, while TDP-43 overexpression did not affect significantly the activity of its promoter (suggesting that TDP-43 does not influence its own transcription), the presence of the 5'UTR sequence and of intron-1 splicing seem to impact positively on TDP-43 expression without affecting transcript stability. In conclusion, we have identified the region spanning nucleotides 451-230 upstream from the transcription start site as the minimal region with a significant transcription activity. These results lay an important foundation for exploring the regulation of the TARDBP gene transcription by exogenous and endogenous stimuli and the implication of transcriptional mechanisms in the pathogenesis of TDP-43 proteinopathies.
Collapse
Affiliation(s)
- Marco Baralle
- grid.425196.d0000 0004 1759 4810International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park, Padriciano, Trieste, Italy
| | - Maurizio Romano
- grid.5133.40000 0001 1941 4308Department of Life Sciences, University of Trieste, Via A. Valerio 28, 34127 Trieste, Italy
| |
Collapse
|
17
|
Rodríguez-Cueto C, Gómez-Almería M, García Toscano L, Romero J, Hillard CJ, de Lago E, Fernández-Ruiz J. Inactivation of the CB 2 receptor accelerated the neuropathological deterioration in TDP-43 transgenic mice, a model of amyotrophic lateral sclerosis. Brain Pathol 2021; 31:e12972. [PMID: 33983653 PMCID: PMC8549023 DOI: 10.1111/bpa.12972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/29/2021] [Accepted: 04/21/2021] [Indexed: 11/29/2022] Open
Abstract
The activation of the cannabinoid receptor type‐2 (CB2) afforded neuroprotection in amyotrophic lateral sclerosis (ALS) models. The objective of this study was to further investigate the relevance of the CB2 receptor through investigating the consequences of its inactivation. TDP‐43(A315T) transgenic mice were crossed with CB2 receptor knock‐out mice to generate double mutants. Temporal and qualitative aspects of the pathological phenotype of the double mutants were compared to TDP‐43 transgenic mice expressing the CB2 receptor. The double mutants exhibited significantly accelerated neurological decline, such that deteriorated rotarod performance was visible at 7 weeks, whereas rotarod performance was normal up to 11 weeks in transgenic mice with intact expression of the CB2 receptor. A morphological analysis of spinal cords confirmed an earlier death (visible at 65 days) of motor neurons labelled with Nissl staining and ChAT immunofluorescence in double mutants compared to TDP‐43 transgenic mice expressing the CB2 receptor. Evidence of glial reactivity, measured using GFAP and Iba‐1 immunostaining, was seen in double mutants at 65 days, but not in TDP‐43 transgenic mice expressing the CB2 receptor. However, at 90 days, both genotypes exhibited similar changes for all these markers, although surviving motor neurons of transgenic mice presented some morphological abnormalities in absence of the CB2 receptor that were not as evident in the presence of this receptor. This faster deterioration seen in double mutants led to premature mortality compared with TDP‐43 transgenic mice expressing the CB2 receptor. We also investigated the consequences of a pharmacological inactivation of the CB2 receptor using the selective antagonist AM630 in TDP‐43 transgenic mice, but results showed only subtle trends towards a greater deterioration. In summary, our results confirmed the potential of the CB2 receptor agonists as a neuroprotective therapy in ALS and strongly support the need to progress towards an evaluation of this potential in patients.
Collapse
Affiliation(s)
- Carmen Rodríguez-Cueto
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Marta Gómez-Almería
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain
| | - Laura García Toscano
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Julián Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eva de Lago
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Javier Fernández-Ruiz
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| |
Collapse
|
18
|
Giunta M, Solje E, Gardoni F, Borroni B, Benussi A. Experimental Disease-Modifying Agents for Frontotemporal Lobar Degeneration. J Exp Pharmacol 2021; 13:359-376. [PMID: 33790662 PMCID: PMC8005747 DOI: 10.2147/jep.s262352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia is a clinically, genetically and pathologically heterogeneous neurodegenerative disorder, enclosing a wide range of different pathological entities, associated with the accumulation of proteins such as tau and TPD-43. Characterized by a high hereditability, mutations in three main genes, MAPT, GRN and C9orf72, can drive the neurodegenerative process. The connection between different genes and proteinopathies through specific mechanisms has shed light on the pathophysiology of the disease, leading to the identification of potential pharmacological targets. New experimental strategies are emerging, in both preclinical and clinical settings, which focus on small molecules rather than gene therapy. In this review, we provide an insight into the aberrant mechanisms leading to FTLD-related proteinopathies and discuss recent therapies with the potential to ameliorate neurodegeneration and disease progression.
Collapse
Affiliation(s)
- Marcello Giunta
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
19
|
Mahdi O, Baharuldin MTH, Nor NHM, Chiroma SM, Jagadeesan S, Moklas MAM. The Neuroprotective Properties, Functions, and Roles of Cannabis sativa in Selected Diseases Related to the Nervous System. Cent Nerv Syst Agents Med Chem 2021; 21:20-38. [PMID: 33504317 DOI: 10.2174/1871524921666210127110028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/17/2020] [Accepted: 12/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cannabis and its extracts are now being explored due to their huge health benefits. Although, the effect they elicit, whether on humans or rodents, may vary based on the age of the animal/subject and or the time in which the extract is administered. However, several debates exist concerning the various medical applications of these compounds. Nonetheless, their applicability as therapeutics should not be clouded based on their perceived negative biological actions. METHODS Articles from reliable databases such as Science Direct, PubMed, Google Scholar, Scopus, and Ovid were searched. Specific search methods were employed using multiple keywords: ''Medicinal Cannabis; endocannabinoid system; cannabinoids receptors; cannabinoids and cognition; brain disorders; neurodegenerative diseases''. For the inclusion/exclusion criteria, only relevant articles related to medicinal Cannabis and its various compounds were considered. RESULTS The current review highlights the role, effects, and involvement of Cannabis, cannabinoids, and endocannabinoids in preventing selected neurodegenerative diseases and possible amelioration of cognitive impairments. Furthermore, it also focuses on Cannabis utilization in many disease conditions such as Alzheimer's and Parkinson's disease among others. CONCLUSION In conclusion, the usage of Cannabis should be further explored as accumulating evidence suggests that it could be effective and somewhat safe, especially when adhered to the recommended dosage. Furthermore, in-depth studies should be conducted in order to unravel the specific mechanism underpinning the involvement of cannabinoids at the cellular level and their therapeutic applications.
Collapse
Affiliation(s)
- Onesimus Mahdi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Selangor, Universiti Putra Malaysia, Sri Serdang 43400, Malaysia
| | - Mohamad T H Baharuldin
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Selangor, Universiti Putra Malaysia, Sri Serdang 43400, Malaysia
| | - Nurul Huda M Nor
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Selangor, Universiti Putra Malaysia, Sri Serdang 43400, Malaysia
| | - Samaila M Chiroma
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Selangor, Universiti Putra Malaysia, Sri Serdang 43400, Malaysia
| | - Saravanan Jagadeesan
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Selangor, Universiti Putra Malaysia, Sri Serdang 43400, Malaysia
| | - Mohamad A M Moklas
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Selangor, Universiti Putra Malaysia, Sri Serdang 43400, Malaysia
| |
Collapse
|
20
|
Pathogenic Genome Signatures That Damage Motor Neurons in Amyotrophic Lateral Sclerosis. Cells 2020; 9:cells9122687. [PMID: 33333804 PMCID: PMC7765192 DOI: 10.3390/cells9122687] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most frequent motor neuron disease and a neurodegenerative disorder, affecting the upper and/or lower motor neurons. Notably, it invariably leads to death within a few years of onset. Although most ALS cases are sporadic, familial amyotrophic lateral sclerosis (fALS) forms 10% of the cases. In 1993, the first causative gene (SOD1) of fALS was identified. With rapid advances in genetics, over fifty potentially causative or disease-modifying genes have been found in ALS so far. Accordingly, routine diagnostic tests should encompass the oldest and most frequently mutated ALS genes as well as several new important genetic variants in ALS. Herein, we discuss current literatures on the four newly identified ALS-associated genes (CYLD, S1R, GLT8D1, and KIF5A) and the previously well-known ALS genes including SOD1, TARDBP, FUS, and C9orf72. Moreover, we review the pathogenic implications and disease mechanisms of these genes. Elucidation of the cellular and molecular functions of the mutated genes will bring substantial insights for the development of therapeutic approaches to treat ALS.
Collapse
|
21
|
de Boer EMJ, Orie VK, Williams T, Baker MR, De Oliveira HM, Polvikoski T, Silsby M, Menon P, van den Bos M, Halliday GM, van den Berg LH, Van Den Bosch L, van Damme P, Kiernan MC, van Es MA, Vucic S. TDP-43 proteinopathies: a new wave of neurodegenerative diseases. J Neurol Neurosurg Psychiatry 2020; 92:jnnp-2020-322983. [PMID: 33177049 PMCID: PMC7803890 DOI: 10.1136/jnnp-2020-322983] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/28/2020] [Accepted: 09/13/2020] [Indexed: 12/31/2022]
Abstract
Inclusions of pathogenic deposits containing TAR DNA-binding protein 43 (TDP-43) are evident in the brain and spinal cord of patients that present across a spectrum of neurodegenerative diseases. For instance, the majority of patients with sporadic amyotrophic lateral sclerosis (up to 97%) and a substantial proportion of patients with frontotemporal lobar degeneration (~45%) exhibit TDP-43 positive neuronal inclusions, suggesting a role for this protein in disease pathogenesis. In addition, TDP-43 inclusions are evident in familial ALS phenotypes linked to multiple gene mutations including the TDP-43 gene coding (TARDBP) and unrelated genes (eg, C9orf72). While TDP-43 is an essential RNA/DNA binding protein critical for RNA-related metabolism, determining the pathophysiological mechanisms through which TDP-43 mediates neurodegeneration appears complex, and unravelling these molecular processes seems critical for the development of effective therapies. This review highlights the key physiological functions of the TDP-43 protein, while considering an expanding spectrum of neurodegenerative diseases associated with pathogenic TDP-43 deposition, and dissecting key molecular pathways through which TDP-43 may mediate neurodegeneration.
Collapse
Affiliation(s)
- Eva Maria Johanna de Boer
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Viyanti K Orie
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Timothy Williams
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mark R Baker
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Clinical Neurophysiology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Hugo M De Oliveira
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Tuomo Polvikoski
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Neuropathology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Matthew Silsby
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Parvathi Menon
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Mehdi van den Bos
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Glenda M Halliday
- Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Leonard H van den Berg
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Philip van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
- Department of Neurology, University Hospital Leuven, Leuven, Belgium
| | - Matthew C Kiernan
- Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Michael A van Es
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Steve Vucic
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Sieverding K, Ulmer J, Bruno C, Satoh T, Tsao W, Freischmidt A, Akira S, Wong PC, Ludolph AC, Danzer KM, Lobsiger CS, Brenner D, Weishaupt JH. Hemizygous deletion of Tbk1 worsens neuromuscular junction pathology in TDP-43 G298S transgenic mice. Exp Neurol 2020; 335:113496. [PMID: 33038415 DOI: 10.1016/j.expneurol.2020.113496] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/26/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022]
Abstract
Mutations in the genes TARDBP (encoding the TDP-43 protein) and TBK1 can cause familial ALS. Neuronal cytoplasmatic accumulations of the misfolded, hyperphosphorylated RNA-binding protein TDP-43 are the pathological hallmark of most ALS cases and have been suggested to be a key aspect of ALS pathogenesis. Pharmacological induction of autophagy has been shown to reduce mutant TDP-43 aggregates and alleviate motor deficits in mice. TBK1 is exemplary for several other ALS genes that regulate autophagy. Consequently, we employed double mutant mice with both a heterozygous Tbk1 deletion and transgenic expression of human TDP-43G298S to test the hypothesis that impaired autophagy reduces intracellular clearance of an aggregation-prone protein and enhances toxicity of mutant TDP-43. The heterozygous deletion of Tbk1 did not change expression or cellular distribution of TDP-43 protein, motor neuron loss or reactive gliosis in the spinal cord of double-mutant mice at the age of 19 months. However, it aggravated muscle denervation and, albeit to a small and variable degree, motor dysfunction in TDP-43G298S transgenic mice, as similarly observed in the SOD1G93A transgenic mouse model for ALS before. Conclusively, our findings suggest that TBK1 mutations can affect the neuromuscular synapse.
Collapse
Affiliation(s)
| | - Johannes Ulmer
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Clara Bruno
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Takashi Satoh
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - William Tsao
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, United States
| | | | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Philip C Wong
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, United States
| | | | - Karin M Danzer
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Christian S Lobsiger
- Institut du Cerveau et de la Moelle Épinière, Institut National de la Santé et de la Recherche Médicale Unité 1127, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7225, Sorbonne Université, Paris, France
| | - David Brenner
- Department of Neurology, University of Ulm, Ulm, Germany; Division of Neurodegenerative Disorders, Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Germany
| | - Jochen H Weishaupt
- Department of Neurology, University of Ulm, Ulm, Germany; Division of Neurodegenerative Disorders, Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Germany.
| |
Collapse
|
23
|
Magalhães Rebelo AP, Dal Bello F, Knedlik T, Kaar N, Volpin F, Shin SH, Giacomello M. Chemical Modulation of Mitochondria-Endoplasmic Reticulum Contact Sites. Cells 2020; 9:cells9071637. [PMID: 32646031 PMCID: PMC7408517 DOI: 10.3390/cells9071637] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Contact sites between mitochondria and endoplasmic reticulum (ER) are points in which the two organelles are in close proximity. Due to their structural and functional complexity, their exploitation as pharmacological targets has never been considered so far. Notwithstanding, the number of compounds described to target proteins residing at these interfaces either directly or indirectly is rising. Here we provide original insight into mitochondria–ER contact sites (MERCs), with a comprehensive overview of the current MERCs pharmacology. Importantly, we discuss the considerable potential of MERCs to become a druggable target for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Ana Paula Magalhães Rebelo
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy; (A.P.M.R.); (F.D.B.); (T.K.); (N.K.); (F.V.); (S.H.S.)
| | - Federica Dal Bello
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy; (A.P.M.R.); (F.D.B.); (T.K.); (N.K.); (F.V.); (S.H.S.)
| | - Tomas Knedlik
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy; (A.P.M.R.); (F.D.B.); (T.K.); (N.K.); (F.V.); (S.H.S.)
| | - Natasha Kaar
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy; (A.P.M.R.); (F.D.B.); (T.K.); (N.K.); (F.V.); (S.H.S.)
| | - Fabio Volpin
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy; (A.P.M.R.); (F.D.B.); (T.K.); (N.K.); (F.V.); (S.H.S.)
| | - Sang Hun Shin
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy; (A.P.M.R.); (F.D.B.); (T.K.); (N.K.); (F.V.); (S.H.S.)
| | - Marta Giacomello
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy; (A.P.M.R.); (F.D.B.); (T.K.); (N.K.); (F.V.); (S.H.S.)
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35121 Padua, Italy
- Correspondence: ; Tel.: +39-049-827-6300
| |
Collapse
|
24
|
Donde A, Sun M, Jeong YH, Wen X, Ling J, Lin S, Braunstein K, Nie S, Wang S, Chen L, Wong PC. Upregulation of ATG7 attenuates motor neuron dysfunction associated with depletion of TARDBP/TDP-43. Autophagy 2020; 16:672-682. [PMID: 31242080 PMCID: PMC7138241 DOI: 10.1080/15548627.2019.1635379] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 05/29/2019] [Accepted: 06/20/2019] [Indexed: 12/12/2022] Open
Abstract
A shared neuropathological hallmark in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is nuclear clearance and cytoplasmic aggregation of TARDBP/TDP-43 (TAR DNA binding protein). We previously showed that the ability of TARDBP to repress nonconserved cryptic exons was impaired in brains of patients with ALS and FTD, suggesting that its nuclear depletion contributes to neurodegeneration. However, the critical pathways impacted by the failure to repress cryptic exons that may contribute to neurodegeneration remain undefined. Here, we report that transcriptome analysis of TARDBP-deficient neurons revealed downregulation of ATG7, a critical gene required for macroautophagy/autophagy. Mouse and Drosophila models lacking TARDBP/TBPH in motor neurons exhibiting age-dependent neurodegeneration and motor deficits showed reduction of ATG7 and accumulation of SQSTM1/p62 inclusions. Importantly, genetic upregulation of the autophagy pathway improved motor function and survival in TBPH-deficient flies. Together with our observation that ATG7 is reduced in ALS-FTD brain tissues, these findings identify the autophagy pathway as one key effector of nuclear depletion of TARDBP that contributes to neurodegeneration. We thus suggest that the autophagy pathway is a therapeutic target for ALS-FTD and other disorders exhibiting TARDBP pathology.Abbreviations: ALS: amyotrophic lateral sclerosis; ANOVA: analysis of variance; ChAT: choline acetyltransferase; CTSD: cathepsin D; FTD: frontotemporal dementia; LAMP1: lysosomal associated membrane protein 1; NMJ: neuromuscular junction; RBFOX3/NeuN: RNA binding fox-1 homolog 3; SQSTM1: sequestosome 1; TARDBP/TDP-43: TAR DNA binding protein 43.
Collapse
Affiliation(s)
- Aneesh Donde
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mingkuan Sun
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yun Ha Jeong
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, Korea
| | - Xinrui Wen
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan Ling
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sophie Lin
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kerstin Braunstein
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shuke Nie
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sheng Wang
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liam Chen
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philip C. Wong
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
25
|
Huang SL, Wu LS, Lee M, Chang CW, Cheng WC, Fang YS, Chen YR, Cheng PL, Shen CKJ. A robust TDP-43 knock-in mouse model of ALS. Acta Neuropathol Commun 2020; 8:3. [PMID: 31964415 PMCID: PMC6975031 DOI: 10.1186/s40478-020-0881-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, adult-onset degenerative disorder of motor neurons. The diseased spinal cord motor neurons of more than 95% of amyotrophic lateral sclerosis (ALS) patients are characterized by the mis-metabolism of the RNA/DNA-binding protein TDP-43 (ALS-TDP), in particular, the presence of cytosolic aggregates of the protein. Most available mouse models for the basic or translational studies of ALS-TDP are based on transgenic overexpression of the TDP-43 protein. Here, we report the generation and characterization of mouse lines bearing homologous knock-in of fALS-associated mutation A315T and sALS-associated mutation N390D, respectively. Remarkably, the heterozygous TDP-43 (N390D/+) mice but not those heterozygous for the TDP-43 (A315T/+) mice develop a full spectrum of ALS-TDP-like pathologies at the molecular, cellular and behavioral levels. Comparative analysis of the mutant mice and spinal cord motor neurons (MN) derived from their embryonic stem (ES) cells demonstrates that different ALS-associated TDP-43 mutations possess critical ALS-causing capabilities and pathogenic pathways, likely modified by their genetic background and the environmental factors. Mechanistically, we identify aberrant RNA splicing of spinal cord Bcl-2 pre-mRNA and consequent increase of a negative regulator of autophagy, Bcl-2, which correlate with and are caused by a progressive increase of TDP-43, one of the early events associated with ALS-TDP pathogenesis, in the spinal cord of TDP-43 (N390D/+) mice and spinal cord MN derived from their ES cells. The TDP-43 (N390D/+) knock-in mice appear to be an ideal rodent model for basic as well as translational studies of ALS- TDP.
Collapse
|
26
|
Filipi T, Hermanova Z, Tureckova J, Vanatko O, Anderova M. Glial Cells-The Strategic Targets in Amyotrophic Lateral Sclerosis Treatment. J Clin Med 2020; 9:E261. [PMID: 31963681 PMCID: PMC7020059 DOI: 10.3390/jcm9010261] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease, which is characterized by the degeneration of motor neurons in the motor cortex and the spinal cord and subsequently by muscle atrophy. To date, numerous gene mutations have been linked to both sporadic and familial ALS, but the effort of many experimental groups to develop a suitable therapy has not, as of yet, proven successful. The original focus was on the degenerating motor neurons, when researchers tried to understand the pathological mechanisms that cause their slow death. However, it was soon discovered that ALS is a complicated and diverse pathology, where not only neurons, but also other cell types, play a crucial role via the so-called non-cell autonomous effect, which strongly deteriorates neuronal conditions. Subsequently, variable glia-based in vitro and in vivo models of ALS were established and used for brand-new experimental and clinical approaches. Such a shift towards glia soon bore its fruit in the form of several clinical studies, which more or less successfully tried to ward the unfavourable prognosis of ALS progression off. In this review, we aimed to summarize current knowledge regarding the involvement of each glial cell type in the progression of ALS, currently available treatments, and to provide an overview of diverse clinical trials covering pharmacological approaches, gene, and cell therapies.
Collapse
Affiliation(s)
- Tereza Filipi
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
- 2nd Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
- 2nd Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| | - Ondrej Vanatko
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, 14200 Prague, Czech Republic; (T.F.); (Z.H.); (J.T.); (O.V.)
| |
Collapse
|
27
|
Kumar V. Endoplasmic Reticulum-Mitochondrial Cross-Talk in Neurodegenerative and Eye Diseases. NEUROLOGY (E-CRONICON) 2019; 11:864-873. [PMID: 31528859 PMCID: PMC6746603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Neurodegenerative diseases demonstrate the progressive decline of brain functions resulting in a significant deterioration in the quality of patient's life. With increasing life expectancy, there has been a significant increase in the incidence of these diseases. Neurodegenerative diseases like Alzheimer's, Parkinson's, and Amyotrophic lateral sclerosis are devastating and afflicts a large world population. Eye, given the similar neural and vascular similarity to the brain, demonstrates many pathological hallmarks of some of these neurological diseases. Moreover, these diseases create an economic and social burden to society. Despite tremendous efforts made in the drug discovery, there is no cure for these fatal diseases. Thus, there is an unmet need to understand cellular and molecular pathophysiology of these diseases. All these diseases demonstrate damage to a large number of seemingly disparate cellular processes and functions such as Ca+2 homeostasis, lipid metabolism, axonal transport, unfolded protein response, autophagy and inflammatory responses. Mitochondria are closely associated with Endoplasmic reticulum (ER) and ER-mitochondrial cross-talk regulates many of these cellular processes and functions damaged in neurodegenerative and eye diseases. Several studies have implicated the disruption of ER-mitochondria contacts in these diseases. This review is aimed at understanding and summarizing the role of ER-mitochondria interacting proteins in major neurodegenerative and eye diseases studied so far.
Collapse
Affiliation(s)
- Varun Kumar
- Department of Ophthalmology, Harvard Medical School, Harvard University, Boston, MA, USA
| |
Collapse
|
28
|
Müller HP, Brenner D, Roselli F, Wiesner D, Abaei A, Gorges M, Danzer KM, Ludolph AC, Tsao W, Wong PC, Rasche V, Weishaupt JH, Kassubek J. Longitudinal diffusion tensor magnetic resonance imaging analysis at the cohort level reveals disturbed cortical and callosal microstructure with spared corticospinal tract in the TDP-43 G298S ALS mouse model. Transl Neurodegener 2019; 8:27. [PMID: 31485326 PMCID: PMC6716821 DOI: 10.1186/s40035-019-0163-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
Background In vivo diffusion tensor imaging (DTI) of the mouse brain was used to identify TDP-43 associated alterations in a mouse model for amyotrophic lateral sclerosis (ALS). Methods Ten mice with TDP-43 G298S overexpression under control of the Thy1.2 promoter and 10 wild type (wt) underwent longitudinal DTI scans at 11.7 T, including one baseline and one follow-up scan with an interval of about 5 months. Whole brain-based spatial statistics (WBSS) of DTI-based parameter maps was used to identify longitudinal alterations of TDP-43 G298S mice compared to wt at the cohort level. Results were supplemented by tractwise fractional anisotropy statistics (TFAS) and histological evaluation of motor cortex for signs of neuronal loss. Results Alterations at the cohort level in TDP-43 G298S mice were observed cross-sectionally and longitudinally in motor areas M1/M2 and in transcallosal fibers but not in the corticospinal tract. Neuronal loss in layer V of motor cortex was detected in TDP-43 G298S at the later (but not at the earlier) timepoint compared to wt. Conclusion DTI mapping of TDP-43 G298S mice demonstrated progression in motor areas M1/M2. WBSS and TFAS are useful techniques to localize TDP-43 G298S associated alterations over time in this ALS mouse model, as a biological marker.
Collapse
Affiliation(s)
- Hans-Peter Müller
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - David Brenner
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Francesco Roselli
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany.,2German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Diana Wiesner
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Alireza Abaei
- 3Core Facility Small Animal MRI, University of Ulm, Ulm, Germany
| | - Martin Gorges
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Karin M Danzer
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Albert C Ludolph
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - William Tsao
- 4Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Philip C Wong
- 4Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Volker Rasche
- 3Core Facility Small Animal MRI, University of Ulm, Ulm, Germany
| | - Jochen H Weishaupt
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Jan Kassubek
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| |
Collapse
|
29
|
Ash PEA, Dhawan U, Boudeau S, Lei S, Carlomagno Y, Knobel M, Al Mohanna LFA, Boomhower SR, Newland MC, Sherr DH, Wolozin B. Heavy Metal Neurotoxicants Induce ALS-Linked TDP-43 Pathology. Toxicol Sci 2019; 167:105-115. [PMID: 30371865 PMCID: PMC6317426 DOI: 10.1093/toxsci/kfy267] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Heavy metals, such as lead, mercury, and selenium, have been epidemiologically linked with a risk of ALS, but a molecular mechanism proving the connection has not been shown. A screen of putative developmental neurotoxins demonstrated that heavy metals (lead, mercury, and tin) trigger accumulation of TDP-43 into nuclear granules with concomitant loss of diffuse nuclear TDP-43. Lead (Pb) and methyl mercury (MeHg) disrupt the homeostasis of TDP-43 in neurons, resulting in increased levels of transcript and increased splicing activity of TDP-43. TDP-43 homeostasis is tightly regulated, and positively or negatively altering its splicing-suppressive activity has been shown to be deleterious to neurons. These changes are associated with the liquid-liquid phase separation of TDP-43 into nuclear bodies. We show that lead directly facilitates phase separation of TDP-43 in a dose-dependent manner in vitro, possibly explaining the means by which lead treatment results in neuronal nuclear granules. Metal toxicants also triggered the accumulation of insoluble TDP-43 in cultured cells and in the cortices of exposed mice. These results provide novel evidence of a direct mechanistic link between heavy metals, which are a commonly cited environmental risk of ALS, and molecular changes in TDP-43, the primary pathological protein accumulating in ALS.
Collapse
Affiliation(s)
- Peter E A Ash
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Uma Dhawan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
- Department of Biomedical Science, Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi 110075, India
| | - Samantha Boudeau
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Shuwen Lei
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Yari Carlomagno
- Neuroscience Division, Mayo Clinic, Jacksonville, Florida 32224
| | - Mark Knobel
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Louloua F A Al Mohanna
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Steven R Boomhower
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115
| | | | - David H Sherr
- Department of Environmental Health, Boston University School of Public Health
| | - Benjamin Wolozin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118
| |
Collapse
|
30
|
Prediger RD, Schamne MG, Sampaio TB, Moreira ELG, Rial D. Animal models of olfactory dysfunction in neurodegenerative diseases. HANDBOOK OF CLINICAL NEUROLOGY 2019; 164:431-452. [PMID: 31604561 DOI: 10.1016/b978-0-444-63855-7.00024-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Olfactory dysfunction seems to occur earlier than classic motor and cognitive symptoms in many neurodegenerative diseases, including Parkinson's disease (PD) and Alzheimer's disease (AD). Thus, the use of the olfactory system as a clinical marker for neurodegenerative diseases is helpful in the characterization of prodromal stages of these diseases, early diagnostic strategies, differential diagnosis, and, potentially, prediction of treatment success. The use of genetic and neurotoxin animal models has contributed to the understanding of the mechanisms underlying olfactory dysfunction in a number of neurodegenerative diseases. In this chapter, we provide an overview of behavioral and neurochemical alterations observed in animal models of different neurodegenerative diseases (such as genetic and Aβ infusion models for AD and neurotoxins and genetic models of PD), in which olfactory dysfunction has been described.
Collapse
Affiliation(s)
- Rui D Prediger
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil.
| | - Marissa G Schamne
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Tuane B Sampaio
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Eduardo L G Moreira
- Department of Physiological Sciences, Center of Biological Sciences¸ Federal University of Santa Catarina, Florianópolis, Brazil
| | - Daniel Rial
- Department of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
31
|
Kefalakes E, Böselt S, Sarikidi A, Ettcheto M, Bursch F, Naujock M, Stanslowsky N, Schmuck M, Barenys M, Wegner F, Grothe C, Petri S. Characterizing the multiple roles of FGF-2 in SOD1 G93A ALS mice in vivo and in vitro. J Cell Physiol 2018; 234:7395-7410. [PMID: 30370540 DOI: 10.1002/jcp.27498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/07/2018] [Indexed: 12/11/2022]
Abstract
We have previously shown that knockout of fibroblast growth factor-2 (FGF-2) and potential compensatory effects of other growth factors result in amelioration of disease symptoms in a transgenic mouse model of amyotrophic lateral sclerosis (ALS). ALS is a rapidly progressive neurological disorder leading to degeneration of cortical, brain stem, and spinal motor neurons followed by subsequent denervation and muscle wasting. Mutations in the superoxide dismutase 1 (SOD1) gene are responsible for approximately 20% of familial ALS cases and SOD1 mutant mice still are among the models best mimicking clinical and neuropathological characteristics of ALS. The aim of the present study was a thorough characterization of FGF-2 and other growth factors and signaling effectors in vivo in the SOD1G93A mouse model. We observed tissue-specific opposing gene regulation of FGF-2 and overall dysregulation of other growth factors, which in the gastrocnemius muscle was associated with reduced downstream extracellular-signal-regulated kinases (ERK) and protein kinase B (AKT) activation. To further investigate whether the effects of FGF-2 on motor neuron death are mediated by glial cells, astrocytes lacking FGF-2 were cocultured together with mutant SOD1 G93A motor neurons. FGF-2 had an impact on motor neuron maturation indicating that astrocytic FGF-2 affects motor neurons at a developmental stage. Moreover, neuronal gene expression patterns showed FGF-2- and SOD1 G93A -dependent changes in ciliary neurotrophic factor, glial-cell-line-derived neurotrophic factor, and ERK2, implying a potential involvement in ALS pathogenesis before the onset of clinical symptoms.
Collapse
Affiliation(s)
- Ekaterini Kefalakes
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Sebastian Böselt
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutical Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Department of Biochemistry, Faculty of Medicine and Life Science, University of Rovira i Virgili, Reus, Spain
| | - Franziska Bursch
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Maximilian Naujock
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Nancy Stanslowsky
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Martin Schmuck
- DAVIS School of Veterinary Medicine, University of California, California
| | - Marta Barenys
- GRET, INSA-UB and Toxicology Unit, Pharmacology, Toxicology and Therapeutical Chemistry Department, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Claudia Grothe
- Center for Systems Neuroscience (ZSN), Hannover, Germany.,Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
32
|
Paul S, Dansithong W, Figueroa KP, Scoles DR, Pulst SM. Staufen1 links RNA stress granules and autophagy in a model of neurodegeneration. Nat Commun 2018; 9:3648. [PMID: 30194296 PMCID: PMC6128856 DOI: 10.1038/s41467-018-06041-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 07/17/2018] [Indexed: 11/26/2022] Open
Abstract
Spinocerebellar ataxia type 2 (SCA2) is a neurodegenerative disease caused by expansion of polyglutamine tract in the ATXN2 protein. We identified Staufen1 (STAU1) as an interactor of ATXN2, and showed elevation in cells from SCA2 patients, amyotrophic lateral sclerosis (ALS) patients, and in SCA2 mouse models. We demonstrated recruitment of STAU1 to mutant ATXN2 aggregates in brain tissue from patients with SCA2 human brain and in an SCA2 mouse model, and association of STAU1 elevation with dysregulation of SCA2-related transcript abundances. Targeting STAU1 in vitro by RNAi restored PCP2 transcript levels and lowering mutant ATXN2 also normalized STAU1 levels. Reduction of Stau1 in vivo improved motor behavior in an SCA2 mouse model, normalized the levels of several SCA2-related proteins, and reduced aggregation of polyglutamine-expanded ATXN2. These findings suggest a function for STAU1 in aberrant RNA metabolism associated with ATXN2 mutation, suggesting STAU1 is a possible novel therapeutic target for SCA2.
Collapse
Affiliation(s)
- Sharan Paul
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA
| | - Warunee Dansithong
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA
| | - Karla P Figueroa
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA
| | - Daniel R Scoles
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, 175 North Medical Drive East, 5th Floor, Salt Lake City, Utah 84132, USA.
| |
Collapse
|
33
|
Ferro D, Yao S, Zarnescu DC. Dynamic duo - FMRP and TDP-43: Regulating common targets, causing different diseases. Brain Res 2018; 1693:37-42. [PMID: 29715444 PMCID: PMC5997554 DOI: 10.1016/j.brainres.2018.04.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 10/17/2022]
Abstract
RNA binding proteins play essential roles during development and aging, and are also involved in disease pathomechanisms. RNA sequencing and omics analyses have provided a window into systems level alterations in neurological disease, and have identified RNA processing defects among notable disease mechanisms. This review focuses on two seemingly distinct neurological disorders, the RNA binding proteins they are linked to, and their newly discovered functional relationship. When deficient, Fragile X Mental Retardation Protein (FMRP) causes developmental deficits and autistic behaviors while TAR-DNA Binding Protein (TDP-43) dysregulation causes age dependent neuronal degeneration. Recent findings that FMRP and TDP-43 associate in ribonuclear protein particles and share mRNA targets in neurons highlight the critical importance of translation regulation in synaptic plasticity and provide new perspectives on neuronal vulnerability during lifespan.
Collapse
Affiliation(s)
- Diana Ferro
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States
| | - Stephen Yao
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States
| | - Daniela C Zarnescu
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States; Department of Neuroscience, University of Arizona, Tucson, AZ, United States; Department of Neurology, University of Arizona, Tucson AZ, United States.
| |
Collapse
|
34
|
Scherz B, Rabl R, Flunkert S, Rohler S, Neddens J, Taub N, Temmel M, Panzenboeck U, Niederkofler V, Zimmermann R, Hutter-Paier B. mTh1 driven expression of hTDP-43 results in typical ALS/FTLD neuropathological symptoms. PLoS One 2018; 13:e0197674. [PMID: 29787578 PMCID: PMC5963763 DOI: 10.1371/journal.pone.0197674] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/07/2018] [Indexed: 12/12/2022] Open
Abstract
Transgenic mouse models are indispensable tools to mimic human diseases and analyze the effectiveness of related new drugs. For a long time amyotrophic lateral sclerosis (ALS) research depended on only a few mouse models that exhibit a very strong and early phenotype, e.g. SOD1 mice, resulting in a short treatment time window. By now, several models are available that need to be characterized to highlight characteristics of each model. Here we further characterized the mThy1-hTDP-43 transgenic mouse model TAR6/6 that overexpresses wild type human TARDBP, also called TDP-43, under control of the neuronal Thy-1 promoter presented by Wils and colleagues, 2010, by using biochemical, histological and behavioral readouts. Our results show that TAR6/6 mice exhibit a strong TDP-43 expression in the hippocampus, spinal cord, hypothalamus and medulla oblongata. Apart from prominent protein expression in the nucleus, TDP-43 protein was found at lower levels in the cytosol of transgenic mice. Additionally, we detected insoluble TDP-43 in the cortex, motoneuron loss, and increased neuroinflammation in the central nervous system of TAR6/6 animals. Behavioral analyses revealed early motor deficits in the clasping- and wire suspension test as well as decreased anxiety in the elevated plus maze. Further motor tests showed differences at later time points compared to non-transgenic littermates, thus allowing the observation of onset and severity of such deficits. Together, TAR6/6 mice are a valuable tool to test new ALS/FTLD drugs that target TDP-43 expression and insolubility, neuroinflammation, motoneuron loss or other TDP-43 related downstream signaling pathways since these mice exhibit a later pathology as previously used ALS/FTLD mouse models.
Collapse
Affiliation(s)
- Barbara Scherz
- QPS Austria GmbH, Grambach, Austria
- Karl-Franzens University, Institute of Molecular Biosciences, Graz, Austria
| | | | | | - Siegfried Rohler
- Medical University Graz, Institute of Pathophysiology and Immunology, Graz, Austria
| | | | | | | | - Ute Panzenboeck
- Medical University Graz, Institute of Pathophysiology and Immunology, Graz, Austria
| | | | - Robert Zimmermann
- Karl-Franzens University, Institute of Molecular Biosciences, Graz, Austria
| | | |
Collapse
|
35
|
Espejo-Porras F, García-Toscano L, Rodríguez-Cueto C, Santos-García I, de Lago E, Fernandez-Ruiz J. Targeting glial cannabinoid CB 2 receptors to delay the progression of the pathological phenotype in TDP-43 (A315T) transgenic mice, a model of amyotrophic lateral sclerosis. Br J Pharmacol 2018; 176:1585-1600. [PMID: 29574689 DOI: 10.1111/bph.14216] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/14/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Cannabinoid CB2 receptors are up-regulated in reactive microglia in the spinal cord of TDP-43 (A315T) transgenic mice, an experimental model of amyotrophic lateral sclerosis. To determine whether this up-regulation can be exploited pharmacologically, we investigated the effects of different treatments that affect CB2 receptor function. EXPERIMENTAL APPROACH We treated TDP-43 (A315T) transgenic mice with the non-selective agonist WIN55,212-2, alone or combined with selective CB1 or CB2 antagonists, as well as with the selective CB2 agonist HU-308, and evaluated their effects on the pathological phenotype. KEY RESULTS WIN55,212-2 had modest beneficial effects in the rotarod test, Nissl staining of motor neurons, and GFAP and Iba-1 immunostainings in the spinal cord, which were mediated in part by CB2 receptor activation. HU-308 significantly improved the rotarod performance of the transgenic mice, with complete preservation of Nissl-stained motor neurons in the ventral horn. Reactive astrogliosis labelled with GFAP was also attenuated by HU-308 in the dorsal and ventral horns, in which CB2 receptors colocalize with this astroglial marker. Furthermore, HU-308 reduced the elevated Iba-1 immunostaining in the ventral horn of TDP-43 transgenic mice, but did not affect this immunoreactivity in white matter, in which CB2 receptors also colocalize with this microglial marker. CONCLUSIONS AND IMPLICATIONS Our study shows an important role for glial CB2 receptors in limiting the progression of the pathological phenotype in TDP-43 (A315T) transgenic mice. Such benefits appear to derive from the activation of CB2 receptors concentrated in astrocytes and reactive microglia located in spinal dorsal and ventral horns. LINKED ARTICLES This article is part of a themed section on 8th European Workshop on Cannabinoid Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.10/issuetoc.
Collapse
Affiliation(s)
- Francisco Espejo-Porras
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Laura García-Toscano
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carmen Rodríguez-Cueto
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Irene Santos-García
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Eva de Lago
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Javier Fernandez-Ruiz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| |
Collapse
|
36
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating, uniformly lethal degenerative disorder of motor neurons that overlaps clinically with frontotemporal dementia (FTD). Investigations of the 10% of ALS cases that are transmitted as dominant traits have revealed numerous gene mutations and variants that either cause these disorders or influence their clinical phenotype. The evolving understanding of the genetic architecture of ALS has illuminated broad themes in the molecular pathophysiology of both familial and sporadic ALS and FTD. These central themes encompass disturbances of protein homeostasis, alterations in the biology of RNA binding proteins, and defects in cytoskeletal dynamics, as well as numerous downstream pathophysiological events. Together, these findings from ALS genetics provide new insight into therapies that target genetically distinct subsets of ALS and FTD.
Collapse
Affiliation(s)
- Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| |
Collapse
|
37
|
Higuchi-Sanabria R, Frankino PA, Paul JW, Tronnes SU, Dillin A. A Futile Battle? Protein Quality Control and the Stress of Aging. Dev Cell 2018; 44:139-163. [PMID: 29401418 PMCID: PMC5896312 DOI: 10.1016/j.devcel.2017.12.020] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/30/2017] [Accepted: 12/20/2017] [Indexed: 12/15/2022]
Abstract
There exists a phenomenon in aging research whereby early life stress can have positive impacts on longevity. The mechanisms underlying these observations suggest a robust, long-lasting induction of cellular defense mechanisms. These include the various unfolded protein responses of the endoplasmic reticulum (ER), cytosol, and mitochondria. Indeed, ectopic induction of these pathways, in the absence of stress, is sufficient to increase lifespan in organisms as diverse as yeast, worms, and flies. Here, we provide an overview of the protein quality control mechanisms that operate in the cytosol, mitochondria, and ER and discuss how they affect cellular health and viability during stress and aging.
Collapse
Affiliation(s)
- Ryo Higuchi-Sanabria
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Phillip Andrew Frankino
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joseph West Paul
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sarah Uhlein Tronnes
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Andrew Dillin
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
38
|
Zhuang J, Wen X, Zhang YQ, Shan Q, Zhang ZF, Zheng GH, Fan SH, Li MQ, Wu DM, Hu B, Lu J, Zheng YL. TDP-43 upregulation mediated by the NLRP3 inflammasome induces cognitive impairment in 2 2',4,4'-tetrabromodiphenyl ether (BDE-47)-treated mice. Brain Behav Immun 2017; 65:99-110. [PMID: 28532818 DOI: 10.1016/j.bbi.2017.05.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/10/2017] [Accepted: 05/19/2017] [Indexed: 10/19/2022] Open
Abstract
It is now commonly known that exposure to polybrominated diphenyl ethers (PBDEs) may cause neurotoxicity and cognitive deficits in children as well as adults, but the underlying mechanisms are still not clear. In the present study, we aimed to elucidate the potential underlying mechanism of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47)-induced neurotoxicity and cognitive impairment. Our results showed that BDE-47-treated mice exhibited impaired cognition and robust upregulation of nuclear TDP-43 in the hippocampus. Hippocampus-specific TDP-43 knockdown attenuated hippocampal apoptosis, restored synaptic protein levels and thus improved cognitive dysfunction in BDE-47-treated mice. Furthermore, our data demonstrated that NLRP3 inflammasome activation played a distinct role in the upregulation of nuclear TDP-43 by downregulating Parkin in the hippocampus of BDE-47-treated mice. Knocking down NLRP3 in the hippocampus or inhibiting caspase 1 activity in BDE-47-treated mice effectively increased Parkin expression in the hippocampus, which decreased the levels of nuclear TDP-43 and ultimately abrogated TDP-43-induced neurotoxic effects. Taken together, our data indicate that TDP-43 upregulation mediated by NLRP3 inflammasome activation via Parkin downregulation in the hippocampus induces cognitive decline in BDE-47-treated mice, and suggest that inhibition of NLRP3 or TDP-43 may be a potential strategy for the prevention or treatment of cognitive impairment in BDE-47-induced neurotoxicity and brain diseases.
Collapse
Affiliation(s)
- Juan Zhuang
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, Jiangsu Province, PR China; Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China; Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, School of Life Science, Huaiyin Normal University, 111 Changjiang Road, Huaian 223300, Jiangsu Province, PR China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China
| | - Yan-Qiu Zhang
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, Jiangsu Province, PR China
| | - Qun Shan
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, Jiangsu Province, PR China; Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China
| | - Gui-Hong Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China
| | - Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China.
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, PR China.
| |
Collapse
|
39
|
Ash PEA, Stanford EA, Al Abdulatif A, Ramirez-Cardenas A, Ballance HI, Boudeau S, Jeh A, Murithi JM, Tripodis Y, Murphy GJ, Sherr DH, Wolozin B. Dioxins and related environmental contaminants increase TDP-43 levels. Mol Neurodegener 2017; 12:35. [PMID: 28476168 PMCID: PMC5420162 DOI: 10.1186/s13024-017-0177-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 04/26/2017] [Indexed: 12/12/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a debilitating neurodegenerative condition that is characterized by progressive loss of motor neurons and the accumulation of aggregated TAR DNA Binding Protein-43 (TDP-43, gene: TARDBP). Increasing evidence indicates that environmental factors contribute to the risk of ALS. Dioxins, related planar polychlorinated biphenyls (PCBs), and polycyclic aromatic hydrocarbons (PAHs) are environmental contaminants that activate the aryl hydrocarbon receptor (AHR), a ligand-activated, PAS family transcription factor. Recently, exposure to these toxicants was identified as a risk factor for ALS. Methods We examined levels of TDP-43 reporter activity, transcript and protein. Quantification was done using cell lines, induced pluripotent stem cells (iPSCs) and mouse brain. The target samples were treated with AHR agonists, including 6-Formylindolo[3,2-b]carbazole (FICZ, a potential endogenous ligand, 2,3,7,8-tetrachlorodibenzo(p)dioxin, and benzo(a)pyrene, an abundant carcinogen in cigarette smoke). The action of the agonists was inhibited by concomitant addition of AHR antagonists or by AHR-specific shRNA. Results We now report that AHR agonists induce up to a 3-fold increase in TDP-43 protein in human neuronal cell lines (BE-M17 cells), motor neuron differentiated iPSCs, and in murine brain. Chronic treatment with AHR agonists elicits over 2-fold accumulation of soluble and insoluble TDP-43, primarily because of reduced TDP-43 catabolism. AHR antagonists or AHR knockdown inhibits agonist-induced increases in TDP-43 protein and TARDBP transcription demonstrating that the ligands act through the AHR. Conclusions These results provide the first evidence that environmental AHR ligands increase TDP-43, which is the principle pathological protein associated with ALS. These results suggest novel molecular mechanisms through which a variety of prevalent environmental factors might directly contribute to ALS. The widespread distribution of dioxins, PCBs and PAHs is considered to be a risk factor for cancer and autoimmune diseases, but could also be a significant public health concern for ALS. Electronic supplementary material The online version of this article (doi:10.1186/s13024-017-0177-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peter E A Ash
- Department of Pharmacology, Boston University School of Medicine, 72 East Concord St., R614, Boston, MA, 02118-2526, USA
| | - Elizabeth A Stanford
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Ali Al Abdulatif
- Department of Pharmacology, Boston University School of Medicine, 72 East Concord St., R614, Boston, MA, 02118-2526, USA
| | | | - Heather I Ballance
- Department of Pharmacology, Boston University School of Medicine, 72 East Concord St., R614, Boston, MA, 02118-2526, USA
| | - Samantha Boudeau
- Department of Pharmacology, Boston University School of Medicine, 72 East Concord St., R614, Boston, MA, 02118-2526, USA
| | - Amanda Jeh
- Department of Pharmacology, Boston University School of Medicine, 72 East Concord St., R614, Boston, MA, 02118-2526, USA
| | - James M Murithi
- Center for Regenerative Medicine, Boston University, Boston, MA, 02118, USA
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - George J Murphy
- Center for Regenerative Medicine, Boston University, Boston, MA, 02118, USA
| | - David H Sherr
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Benjamin Wolozin
- Department of Pharmacology, Boston University School of Medicine, 72 East Concord St., R614, Boston, MA, 02118-2526, USA. .,Department of Neurology, Boston University School of Medicine, 72 East Concord St., R614, Boston, MA, 02118-2526, USA.
| |
Collapse
|
40
|
Mouse models of frontotemporal dementia: A comparison of phenotypes with clinical symptomatology. Neurosci Biobehav Rev 2017; 74:126-138. [DOI: 10.1016/j.neubiorev.2017.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 12/12/2022]
|
41
|
Jeong YH, Ling JP, Lin SZ, Donde AN, Braunstein KE, Majounie E, Traynor BJ, LaClair KD, Lloyd TE, Wong PC. Tdp-43 cryptic exons are highly variable between cell types. Mol Neurodegener 2017; 12:13. [PMID: 28153034 PMCID: PMC5289002 DOI: 10.1186/s13024-016-0144-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 12/20/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND TDP-43 proteinopathy is a prominent pathological feature that occurs in a number of human diseases including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and inclusion body myositis (IBM). Our recent finding that TDP-43 represses nonconserved cryptic exons led us to ask whether cell type-specific cryptic exons could exist to impact unique molecular pathways in brain or muscle. METHODS In the present work, we investigated TDP-43's function in various mouse tissues to model disease pathogenesis. We generated mice to conditionally delete TDP-43 in excitatory neurons or skeletal myocytes and identified the cell type-specific cryptic exons associated with TDP-43 loss of function. RESULTS Comparative analysis of nonconserved cryptic exons in various mouse cell types revealed that only some cryptic exons were common amongst stem cells, neurons, and myocytes; the majority of these nonconserved cryptic exons were cell type-specific. CONCLUSIONS Our results suggest that in human disease, TDP-43 loss of function may impair cell type-specific pathways.
Collapse
Affiliation(s)
- Yun Ha Jeong
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Neural Development and Disease Department, Korea Brain Research Institute, Daegu, 701-300 South Korea
| | - Jonathan P. Ling
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Sophie Z. Lin
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Aneesh N. Donde
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Kerstin E. Braunstein
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Elisa Majounie
- Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD 20892 USA
- Present address: Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Cardiff, CF24 4HQ UK
| | - Bryan J. Traynor
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Laboratory of Neurogenetics, NIA, NIH, Bethesda, MD 20892 USA
| | - Katherine D. LaClair
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Thomas E. Lloyd
- Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Philip C. Wong
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
42
|
Rauskolb S, Dombert B, Sendtner M. Insulin-like growth factor 1 in diabetic neuropathy and amyotrophic lateral sclerosis. Neurobiol Dis 2017; 97:103-113. [DOI: 10.1016/j.nbd.2016.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/29/2016] [Accepted: 04/29/2016] [Indexed: 12/12/2022] Open
|
43
|
Suryadevara U, Bruijnzeel DM, Nuthi M, Jagnarine DA, Tandon R, Bruijnzeel AW. Pros and Cons of Medical Cannabis use by People with Chronic Brain Disorders. Curr Neuropharmacol 2017; 15:800-814. [PMID: 27804883 PMCID: PMC5652027 DOI: 10.2174/1570159x14666161101095325] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 08/26/2016] [Accepted: 10/28/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cannabis is the most widely used illicit drug in the world and there is growing concern about the mental health effects of cannabis use. These concerns are at least partly due to the strong increase in recreational and medical cannabis use and the rise in tetrahydrocannabinol (THC) levels. Cannabis is widely used to self-medicate by older people and people with brain disorders such as amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), bipolar disorder, and schizophrenia. OBJECTIVE This review provides an overview of the perceived benefits and adverse mental health effects of cannabis use in people with ALS, MS, AD, PD, bipolar disorder, and schizophrenia. RESULTS The reviewed studies indicate that cannabis use diminishes some symptoms associated with these disorders. Cannabis use decreases pain and spasticity in people with MS, decreases tremor, rigidity, and pain in people with PD, and improves the quality of life of ALS patients by improving appetite, and decreasing pain and spasticity. Cannabis use is more common among people with schizophrenia than healthy controls. Cannabis use is a risk factor for schizophrenia which increases positive symptoms in schizophrenia patients and diminishes negative symptoms. Cannabis use worsens bipolar disorder and there is no evidence that bipolar patients derive any benefit from cannabis. In late stage Alzheimer's patients, cannabis products may improve food intake, sleep quality, and diminish agitation. CONCLUSION Cannabis use diminishes some of the adverse effects of neurological and psychiatric disorders. However, chronic cannabis use may lead to cognitive impairments and dependence.
Collapse
Affiliation(s)
- Uma Suryadevara
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | | | - Meena Nuthi
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | | | - Rajiv Tandon
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Adriaan W. Bruijnzeel
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, Florida, USA
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| |
Collapse
|
44
|
Alfieri JA, Silva PR, Igaz LM. Early Cognitive/Social Deficits and Late Motor Phenotype in Conditional Wild-Type TDP-43 Transgenic Mice. Front Aging Neurosci 2016; 8:310. [PMID: 28066234 PMCID: PMC5167738 DOI: 10.3389/fnagi.2016.00310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/06/2016] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal Dementia (FTD) and amyotrophic lateral sclerosis (ALS) are two neurodegenerative diseases associated to mislocalization and aggregation of TAR DNA-binding protein 43 (TDP-43). To investigate in depth the behavioral phenotype associated with this proteinopathy, we used as a model transgenic (Tg) mice conditionally overexpressing human wild-type TDP 43 protein (hTDP-43-WT) in forebrain neurons. We previously characterized these mice at the neuropathological level and found progressive neurodegeneration and other features that evoke human TDP-43 proteinopathies of the FTD/ALS spectrum. In the present study we analyzed the behavior of mice at multiple domains, including motor, social and cognitive performance. Our results indicate that young hTDP-43-WT Tg mice (1 month after post-weaning transgene induction) present a normal motor phenotype compared to control littermates, as assessed by accelerated rotarod performance, spontaneous locomotor activity in the open field test and a mild degree of spasticity shown by a clasping phenotype. Analysis of social and cognitive behavior showed a rapid installment of deficits in social interaction, working memory (Y-maze test) and recognition memory (novel object recognition test) in the absence of overt motor abnormalities. To investigate if the motor phenotype worsen with age, we analyzed the behavior of mice after long-term (up to 12 months) transgene induction. Our results reveal a decreased performance on the rotarod test and in the hanging wire test, indicating a motor phenotype that was absent in younger mice. In addition, long-term hTDP-43-WT expression led to hyperlocomotion in the open field test. In sum, these results demonstrate a time-dependent emergence of a motor phenotype in older hTDP-43-WT Tg mice, recapitulating aspects of clinical FTD presentations with motor involvement in human patients, and providing a complementary animal model for studying TDP-43 proteinopathies.
Collapse
Affiliation(s)
- Julio A Alfieri
- IFIBIO Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires - CONICET Buenos Aires, Argentina
| | - Pablo R Silva
- IFIBIO Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires - CONICET Buenos Aires, Argentina
| | - Lionel M Igaz
- IFIBIO Houssay, Grupo de Neurociencia de Sistemas, Facultad de Medicina, Universidad de Buenos Aires - CONICET Buenos Aires, Argentina
| |
Collapse
|
45
|
TDP-43 aggregation mirrors TDP-43 knockdown, affecting the expression levels of a common set of proteins. Sci Rep 2016; 6:33996. [PMID: 27665936 PMCID: PMC5036055 DOI: 10.1038/srep33996] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/06/2016] [Indexed: 12/12/2022] Open
Abstract
TDP-43 protein plays an important role in regulating transcriptional repression, RNA metabolism, and splicing. Typically it shuttles between the nucleus and the cytoplasm to perform its functions, while abnormal cytoplasmic aggregation of TDP-43 has been associated with neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). For the purpose of this study we selected a set of proteins that were misregulated following silencing of TDP-43 and analysed their expression in a TDP-43-aggregation model cell line HEK293 Flp-in Flag-TDP-43-12x-Q/N F4L. Following TDP-43 sequestration in insoluble aggregates, we observed higher nuclear levels of EIF4A3, and POLDIP3β, whereas nuclear levels of DNMT3A, HNRNPA3, PABPC1 and POLDIP3α dropped, and cytoplasmic levels of RANBP1 dropped. In addition, immunofluorescence signal intensity quantifications showed increased nuclear expression of HNRNPL and YARS, and downregulation of cytoplasmic DPCD. Furthermore, cytoplasmic levels of predominantly nuclear protein ALYREF increased. In conclusion, by identifying a common set of proteins that are differentially expressed in a similar manner in these two different conditions, we show that TDP-43 aggregation has a comparable effect to TDP-43 knockdown.
Collapse
|
46
|
Cookson MR. RNA-binding proteins implicated in neurodegenerative diseases. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 8. [PMID: 27659605 DOI: 10.1002/wrna.1397] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/23/2016] [Accepted: 08/27/2016] [Indexed: 12/12/2022]
Abstract
Gene expression is regulated at many levels, including after generation of the primary RNA transcript from DNA but before translation into protein. Such post-translational gene regulation occurs via the action of a multitude of RNA binding proteins and include varied actions from splicing to regulation of association with the translational machinery. Primary evidence that such processes might contribute to disease mechanisms in neurodegenerative disorders comes from the observation of mutations in RNA binding proteins, particularly in diseases in the amyotrophic lateral sclerosis-frontotemporal dementia spectrum and in some forms of ataxia and tremor. The bulk of evidence from recent surveys of the types of RNA species that are affected in these disorders suggests a global deregulation of control rather than a very small number of RNA species, although why some groups of neurons are sensitive to these changes is not well understood. Overall, these data suggest that neurodegeneration can be initiated by mutations in RNA binding proteins and, as a corollary, that neurons are particularly sensitive to loss of control of gene expression at the post-transcriptional level. Such observations have implications not only for understanding the nature of neurodegenerative disorders but also how we might intervene therapeutically in these diseases. WIREs RNA 2017, 8:e1397. doi: 10.1002/wrna.1397 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
47
|
Stoica R, Paillusson S, Gomez-Suaga P, Mitchell JC, Lau DH, Gray EH, Sancho RM, Vizcay-Barrena G, De Vos KJ, Shaw CE, Hanger DP, Noble W, Miller CC. ALS/FTD-associated FUS activates GSK-3β to disrupt the VAPB-PTPIP51 interaction and ER-mitochondria associations. EMBO Rep 2016; 17:1326-42. [PMID: 27418313 PMCID: PMC5007559 DOI: 10.15252/embr.201541726] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 05/06/2016] [Accepted: 06/13/2016] [Indexed: 12/12/2022] Open
Abstract
Defective FUS metabolism is strongly associated with amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), but the mechanisms linking FUS to disease are not properly understood. However, many of the functions disrupted in ALS/FTD are regulated by signalling between the endoplasmic reticulum (ER) and mitochondria. This signalling is facilitated by close physical associations between the two organelles that are mediated by binding of the integral ER protein VAPB to the outer mitochondrial membrane protein PTPIP51, which act as molecular scaffolds to tether the two organelles. Here, we show that FUS disrupts the VAPB-PTPIP51 interaction and ER-mitochondria associations. These disruptions are accompanied by perturbation of Ca(2+) uptake by mitochondria following its release from ER stores, which is a physiological read-out of ER-mitochondria contacts. We also demonstrate that mitochondrial ATP production is impaired in FUS-expressing cells; mitochondrial ATP production is linked to Ca(2+) levels. Finally, we demonstrate that the FUS-induced reductions to ER-mitochondria associations and are linked to activation of glycogen synthase kinase-3β (GSK-3β), a kinase already strongly associated with ALS/FTD.
Collapse
Affiliation(s)
- Radu Stoica
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Sébastien Paillusson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Patricia Gomez-Suaga
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Jacqueline C Mitchell
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Dawn Hw Lau
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Emma H Gray
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Rosa M Sancho
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | | | - Kurt J De Vos
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Christopher E Shaw
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Diane P Hanger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Christopher Cj Miller
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| |
Collapse
|
48
|
Blasco H, Patin F, Andres CR, Corcia P, Gordon PH. Amyotrophic Lateral Sclerosis, 2016: existing therapies and the ongoing search for neuroprotection. Expert Opin Pharmacother 2016; 17:1669-82. [PMID: 27356036 DOI: 10.1080/14656566.2016.1202919] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS), one in a family of age-related neurodegenerative disorders, is marked by predominantly cryptogenic causes, partially elucidated pathophysiology, and elusive treatments. The challenges of ALS are illustrated by two decades of negative drug trials. AREAS COVERED In this article, we lay out the current understanding of disease genesis and physiology in relation to drug development in ALS, stressing important accomplishments and gaps in knowledge. We briefly consider clinical ALS, the ongoing search for biomarkers, and the latest in trial design, highlighting major recent and ongoing clinical trials; and we discuss, in a concluding section on future directions, the prion-protein hypothesis of neurodegeneration and what steps can be taken to end the drought that has characterized drug discovery in ALS. EXPERT OPINION Age-related neurodegenerative disorders are fast becoming major public health problems for the world's aging populations. Several agents offer promise in the near-term, but drug development is hampered by an interrelated cycle of obstacles surrounding etiological, physiological, and biomarkers discovery. It is time for the type of government-funded, public-supported offensive on neurodegenerative disease that has been effective in other fields.
Collapse
Affiliation(s)
- H Blasco
- a Inserm U930, Equipe "neurogénétique et neurométabolomique" , Tours , France.,b Université François-Rabelais, Faculté de Médecine , Tours , France.,c Laboratoire de Biochimie et Biologie Moléculaire , CHRU de Tours , Tours , France
| | - F Patin
- a Inserm U930, Equipe "neurogénétique et neurométabolomique" , Tours , France.,b Université François-Rabelais, Faculté de Médecine , Tours , France.,c Laboratoire de Biochimie et Biologie Moléculaire , CHRU de Tours , Tours , France
| | - C R Andres
- a Inserm U930, Equipe "neurogénétique et neurométabolomique" , Tours , France.,b Université François-Rabelais, Faculté de Médecine , Tours , France.,c Laboratoire de Biochimie et Biologie Moléculaire , CHRU de Tours , Tours , France
| | - P Corcia
- a Inserm U930, Equipe "neurogénétique et neurométabolomique" , Tours , France.,b Université François-Rabelais, Faculté de Médecine , Tours , France.,d Centre SLA, Service de Neurologie , CHRU Bretonneau , Tours , France
| | - P H Gordon
- e Northern Navajo Medical Center , Neurology Unit , Shiprock , NM , USA
| |
Collapse
|
49
|
Akamatsu M, Yamashita T, Hirose N, Teramoto S, Kwak S. The AMPA receptor antagonist perampanel robustly rescues amyotrophic lateral sclerosis (ALS) pathology in sporadic ALS model mice. Sci Rep 2016; 6:28649. [PMID: 27350567 PMCID: PMC4923865 DOI: 10.1038/srep28649] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/06/2016] [Indexed: 02/08/2023] Open
Abstract
Both TDP-43 pathology and failure of RNA editing of AMPA receptor subunit GluA2, are etiology-linked molecular abnormalities that concomitantly occur in the motor neurons of the majority of patients with amyotrophic lateral sclerosis (ALS). AR2 mice, in which an RNA editing enzyme adenosine deaminase acting on RNA 2 (ADAR2) is conditionally knocked out in the motor neurons, exhibit a progressive ALS phenotype with TDP-43 pathology in the motor neurons through a Ca2+-permeable AMPA receptor-mediated mechanism. Therefore, amelioration of the increased Ca2+ influx by AMPA receptor antagonists may be a potential ALS therapy. Here, we showed that orally administered perampanel, a selective, non-competitive AMPA receptor antagonist significantly prevented the progression of the ALS phenotype and normalized the TDP-43 pathology-associated death of motor neurons in the AR2 mice. Given that perampanel is an approved anti-epileptic drug, perampanel is a potential candidate ALS drug worthy of a clinical trial.
Collapse
Affiliation(s)
- Megumi Akamatsu
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takenari Yamashita
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Naoki Hirose
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sayaka Teramoto
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shin Kwak
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Clinical Research Center for Medicine, International University of Health and Welfare, Ichikawa, Chiba, Japan
| |
Collapse
|
50
|
Koyama A, Sugai A, Kato T, Ishihara T, Shiga A, Toyoshima Y, Koyama M, Konno T, Hirokawa S, Yokoseki A, Nishizawa M, Kakita A, Takahashi H, Onodera O. Increased cytoplasmic TARDBP mRNA in affected spinal motor neurons in ALS caused by abnormal autoregulation of TDP-43. Nucleic Acids Res 2016; 44:5820-36. [PMID: 27257061 PMCID: PMC4937342 DOI: 10.1093/nar/gkw499] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 05/23/2016] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disorder. In motor neurons of ALS, TAR DNA binding protein-43 (TDP-43), a nuclear protein encoded by TARDBP, is absent from the nucleus and forms cytoplasmic inclusions. TDP-43 auto-regulates the amount by regulating the TARDBP mRNA, which has three polyadenylation signals (PASs) and three additional alternative introns within the last exon. However, it is still unclear how the autoregulatory mechanism works and how the status of autoregulation in ALS motor neurons without nuclear TDP-43 is. Here we show that TDP-43 inhibits the selection of the most proximal PAS and induces splicing of multiple alternative introns in TARDBP mRNA to decrease the amount of cytoplasmic TARDBP mRNA by nonsense-mediated mRNA decay. When TDP-43 is depleted, the TARDBP mRNA uses the most proximal PAS and is increased in the cytoplasm. Finally, we have demonstrated that in ALS motor neurons—especially neurons with mislocalized TDP-43—the amount of TARDBP mRNA is increased in the cytoplasm. Our observations indicate that nuclear TDP-43 contributes to the autoregulation and suggests that the absence of nuclear TDP-43 induces an abnormal autoregulation and increases the amount of TARDBP mRNA. The vicious cycle might accelerate the disease progression of ALS.
Collapse
Affiliation(s)
- Akihide Koyama
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan Center for Transdisciplinary Research, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Akihiro Sugai
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Taisuke Kato
- Department of Molecular Neuroscience, Resource Branch for Brain Disease Research, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Tomohiko Ishihara
- Department of Molecular Neuroscience, Resource Branch for Brain Disease Research, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Atsushi Shiga
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan Center for Transdisciplinary Research, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Yasuko Toyoshima
- Department of Pathology, Clinical Neuroscience Branch, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Misaki Koyama
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Takuya Konno
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Sachiko Hirokawa
- Department of Molecular Neuroscience, Resource Branch for Brain Disease Research, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Akio Yokoseki
- Department of Molecular Neuroscience, Resource Branch for Brain Disease Research, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Masatoyo Nishizawa
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Center for Bioresource-based Research, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Hitoshi Takahashi
- Department of Pathology, Clinical Neuroscience Branch, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| | - Osamu Onodera
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, 1-757 Asahimachi-dori, Chuo-ku, Niigata-City, Niigata 951-8585, Japan
| |
Collapse
|