1
|
Kim S, Phan S, Tran HT, Shaw TR, Shahmoradian SH, Ellisman MH, Veatch SL, Barmada SJ, Pappas SS, Dauer WT. TorsinA is essential for neuronal nuclear pore complex localization and maturation. Nat Cell Biol 2024; 26:1482-1495. [PMID: 39117796 DOI: 10.1038/s41556-024-01480-1] [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: 04/15/2023] [Accepted: 07/11/2024] [Indexed: 08/10/2024]
Abstract
As lifelong interphase cells, neurons face an array of unique challenges. A key challenge is regulating nuclear pore complex (NPC) biogenesis and localization, the mechanisms of which are largely unknown. Here we identify neuronal maturation as a period of strongly upregulated NPC biogenesis. We demonstrate that the AAA+ protein torsinA, whose dysfunction causes the neurodevelopmental movement disorder DYT-TOR1A dystonia and co-ordinates NPC spatial organization without impacting total NPC density. We generated an endogenous Nup107-HaloTag mouse line to directly visualize NPC organization in developing neurons and find that torsinA is essential for proper NPC localization. In the absence of torsinA, the inner nuclear membrane buds excessively at sites of mislocalized nascent NPCs, and the formation of complete NPCs is delayed. Our work demonstrates that NPC spatial organization and number are independently determined and identifies NPC biogenesis as a process vulnerable to neurodevelopmental disease insults.
Collapse
Affiliation(s)
- Sumin Kim
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sébastien Phan
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Hung Tri Tran
- Peter O'Donnell Jr. Brain Institute, UT Southwestern, Dallas, TX, USA
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern, Dallas, TX, USA
| | - Thomas R Shaw
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
- Program in Applied Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Sarah H Shahmoradian
- Peter O'Donnell Jr. Brain Institute, UT Southwestern, Dallas, TX, USA
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern, Dallas, TX, USA
- Department of Biophysics, UT Southwestern, Dallas, TX, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sarah L Veatch
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
- Program in Applied Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Sami J Barmada
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI, USA.
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| | - Samuel S Pappas
- Peter O'Donnell Jr. Brain Institute, UT Southwestern, Dallas, TX, USA.
- Department of Neurology, UT Southwestern, Dallas, TX, USA.
| | - William T Dauer
- Peter O'Donnell Jr. Brain Institute, UT Southwestern, Dallas, TX, USA.
- Department of Neurology, UT Southwestern, Dallas, TX, USA.
- Department of Neuroscience, UT Southwestern, Dallas, TX, USA.
| |
Collapse
|
2
|
Wolf D, Ayon-Olivas M, Sendtner M. BDNF-Regulated Modulation of Striatal Circuits and Implications for Parkinson's Disease and Dystonia. Biomedicines 2024; 12:1761. [PMID: 39200225 PMCID: PMC11351984 DOI: 10.3390/biomedicines12081761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Neurotrophins, particularly brain-derived neurotrophic factor (BDNF), act as key regulators of neuronal development, survival, and plasticity. BDNF is necessary for neuronal and functional maintenance in the striatum and the substantia nigra, both structures involved in the pathogenesis of Parkinson's Disease (PD). Depletion of BDNF leads to striatal degeneration and defects in the dendritic arborization of striatal neurons. Activation of tropomyosin receptor kinase B (TrkB) by BDNF is necessary for the induction of long-term potentiation (LTP), a form of synaptic plasticity, in the hippocampus and striatum. PD is characterized by the degeneration of nigrostriatal neurons and altered striatal plasticity has been implicated in the pathophysiology of PD motor symptoms, leading to imbalances in the basal ganglia motor pathways. Given its essential role in promoting neuronal survival and meditating synaptic plasticity in the motor system, BDNF might have an important impact on the pathophysiology of neurodegenerative diseases, such as PD. In this review, we focus on the role of BDNF in corticostriatal plasticity in movement disorders, including PD and dystonia. We discuss the mechanisms of how dopaminergic input modulates BDNF/TrkB signaling at corticostriatal synapses and the involvement of these mechanisms in neuronal function and synaptic plasticity. Evidence for alterations of BDNF and TrkB in PD patients and animal models are reviewed, and the potential of BDNF to act as a therapeutic agent is highlighted. Advancing our understanding of these mechanisms could pave the way toward innovative therapeutic strategies aiming at restoring neuroplasticity and enhancing motor function in these diseases.
Collapse
Affiliation(s)
| | | | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078 Wuerzburg, Germany (M.A.-O.)
| |
Collapse
|
3
|
Lewis SA, Forstrom J, Tavani J, Schafer R, Tiede Z, Padilla-Lopez SR, Kruer MC. eIF2α phosphorylation evokes dystonia-like movements with D2-receptor and cholinergic origin and abnormal neuronal connectivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594240. [PMID: 38798458 PMCID: PMC11118466 DOI: 10.1101/2024.05.14.594240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Dystonia is the 3rd most common movement disorder. Dystonia is acquired through either injury or genetic mutations, with poorly understood molecular and cellular mechanisms. Eukaryotic initiation factor alpha (eIF2α) controls cell state including neuronal plasticity via protein translation control and expression of ATF4. Dysregulated eIF2α phosphorylation (eIF2α-P) occurs in dystonia patients and models including DYT1, but the consequences are unknown. We increased/decreased eIF2α-P and tested motor control and neuronal properties in a Drosophila model. Bidirectionally altering eIF2α-P produced dystonia-like abnormal posturing and dyskinetic movements in flies. These movements were also observed with expression of the DYT1 risk allele. We identified cholinergic and D2-receptor neuroanatomical origins of these dyskinetic movements caused by genetic manipulations to dystonia molecular candidates eIF2α-P, ATF4, or DYT1, with evidence for decreased cholinergic release. In vivo, increased and decreased eIF2α-P increase synaptic connectivity at the NMJ with increased terminal size and bouton synaptic release sites. Long-term treatment of elevated eIF2α-P with ISRIB restored adult longevity, but not performance in a motor assay. Disrupted eIF2α-P signaling may alter neuronal connectivity, change synaptic release, and drive motor circuit changes in dystonia.
Collapse
Affiliation(s)
- Sara A Lewis
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Jacob Forstrom
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Jennifer Tavani
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Robert Schafer
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Zach Tiede
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Sergio R Padilla-Lopez
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
| | - Michael C Kruer
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA
- Departments of Child Health, Cellular & Molecular Medicine, Genetics, and Neurology, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA
- Programs in Neuroscience, Molecular & Cellular Biology, and Biomedical Informatics, Arizona State University, Tempe, AZ USA
| |
Collapse
|
4
|
Akter M, Cui H, Hosain MA, Liu J, Duan Y, Ding B. RANBP17 Overexpression Restores Nucleocytoplasmic Transport and Ameliorates Neurodevelopment in Induced DYT1 Dystonia Motor Neurons. J Neurosci 2024; 44:e1728232024. [PMID: 38438257 PMCID: PMC11007476 DOI: 10.1523/jneurosci.1728-23.2024] [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: 09/11/2023] [Revised: 02/18/2024] [Accepted: 02/20/2024] [Indexed: 03/06/2024] Open
Abstract
DYT1 dystonia is a debilitating neurological movement disorder, and it represents the most frequent and severe form of hereditary primary dystonia. There is currently no cure for this disease due to its unclear pathogenesis. In our previous study utilizing patient-specific motor neurons (MNs), we identified distinct cellular deficits associated with the disease, including a deformed nucleus, disrupted neurodevelopment, and compromised nucleocytoplasmic transport (NCT) functions. However, the precise molecular mechanisms underlying these cellular impairments have remained elusive. In this study, we revealed the genome-wide changes in gene expression in DYT1 MNs through transcriptomic analysis. We found that those dysregulated genes are intricately involved in neurodevelopment and various biological processes. Interestingly, we identified that the expression level of RANBP17, a RAN-binding protein crucial for NCT regulation, exhibited a significant reduction in DYT1 MNs. By manipulating RANBP17 expression, we further demonstrated that RANBP17 plays an important role in facilitating the nuclear transport of both protein and transcript cargos in induced human neurons. Excitingly, the overexpression of RANBP17 emerged as a substantial mitigating factor, effectively restoring impaired NCT activity and rescuing neurodevelopmental deficits observed in DYT1 MNs. These findings shed light on the intricate molecular underpinnings of impaired NCT in DYT1 neurons and provide novel insights into the pathophysiology of DYT1 dystonia, potentially leading to the development of innovative treatment strategies.
Collapse
Affiliation(s)
- Masuma Akter
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, Louisiana 71130-3932
| | - Haochen Cui
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, Louisiana 71130-3932
| | - Md Abir Hosain
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, Louisiana 71130-3932
| | - Jinmei Liu
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, Louisiana 71130-3932
| | - Yuntian Duan
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, Louisiana 71130-3932
| | - Baojin Ding
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, Louisiana 71130-3932
| |
Collapse
|
5
|
Pan K, Jinnah HA, Hess EJ, Smith Y, Villalba RM. Ultrastructural analysis of nigrostriatal dopaminergic terminals in a knockin mouse model of DYT1 dystonia. Eur J Neurosci 2024; 59:1407-1427. [PMID: 38123503 DOI: 10.1111/ejn.16197] [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: 03/08/2023] [Revised: 11/02/2023] [Accepted: 11/05/2023] [Indexed: 12/23/2023]
Abstract
DYT1 dystonia is associated with decreased striatal dopamine release. In this study, we examined the possibility that ultrastructural changes of nigrostriatal dopamine terminals could contribute to this neurochemical imbalance using a serial block face/scanning electron microscope (SBF/SEM) and three-dimensional reconstruction to analyse striatal tyrosine hydroxylase-immunoreactive (TH-IR) terminals and their synapses in a DYT1(ΔE) knockin (DYT1-KI) mouse model of DYT1 dystonia. Furthermore, to study possible changes in vesicle packaging capacity of dopamine, we used transmission electron microscopy to assess the synaptic vesicle size in striatal dopamine terminals. Quantitative comparative analysis of 80 fully reconstructed TH-IR terminals in the WT and DYT1-KI mice indicate (1) no significant difference in the volume of TH-IR terminals; (2) no major change in the proportion of axo-spinous versus axo-dendritic synapses; (3) no significant change in the post-synaptic density (PSD) area of axo-dendritic synapses, while the PSDs of axo-spinous synapses were significantly smaller in DYT1-KI mice; (4) no significant change in the contact area between TH-IR terminals and dendritic shafts or spines, while the ratio of PSD area/contact area decreased significantly for both axo-dendritic and axo-spinous synapses in DYT1-KI mice; (5) no significant difference in the mitochondria volume; and (6) no significant difference in the synaptic vesicle area between the two groups. Altogether, these findings suggest that abnormal morphometric changes of nigrostriatal dopamine terminals and their post-synaptic targets are unlikely to be a major source of reduced striatal dopamine release in DYT1 dystonia.
Collapse
Affiliation(s)
- Ke Pan
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Physical Therapy & Human Movement Sciences, Northwestern University, Chicago, Illinois, USA
| | - Hyder A Jinnah
- Department of Neurology, Emory University, Atlanta, Georgia, USA
- Department of Human Genetics and Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Ellen J Hess
- Department of Neurology, Emory University, Atlanta, Georgia, USA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, Georgia, USA
| | - Yoland Smith
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Rosa M Villalba
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
6
|
Peall KJ, Owen MJ, Hall J. Rare genetic brain disorders with overlapping neurological and psychiatric phenotypes. Nat Rev Neurol 2024; 20:7-21. [PMID: 38001363 DOI: 10.1038/s41582-023-00896-x] [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] [Accepted: 10/27/2023] [Indexed: 11/26/2023]
Abstract
Understanding rare genetic brain disorders with overlapping neurological and psychiatric phenotypes is of increasing importance given the potential for developing disease models that could help to understand more common, polygenic disorders. However, the traditional clinical boundaries between neurology and psychiatry result in frequent segregation of these disorders into distinct silos, limiting cross-specialty understanding that could facilitate clinical and biological advances. In this Review, we highlight multiple genetic brain disorders in which neurological and psychiatric phenotypes are observed, but for which in-depth, cross-spectrum clinical phenotyping is rarely undertaken. We describe the combined phenotypes observed in association with genetic variants linked to epilepsy, dystonia, autism spectrum disorder and schizophrenia. We also consider common underlying mechanisms that centre on synaptic plasticity, including changes to synaptic and neuronal structure, calcium handling and the balance of excitatory and inhibitory neuronal activity. Further investigation is needed to better define and replicate these phenotypes in larger cohorts, which would help to gain greater understanding of the pathophysiological mechanisms and identify common therapeutic targets.
Collapse
Affiliation(s)
- Kathryn J Peall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK.
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.
| | - Michael J Owen
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
7
|
García-García E, Ramón-Lainez A, Conde-Berriozabal S, Del Toro D, Escaramis G, Giralt A, Masana M, Alberch J, Rodríguez MJ. VPS13A knockdown impairs corticostriatal synaptic plasticity and locomotor behavior in a new mouse model of chorea-acanthocytosis. Neurobiol Dis 2023; 187:106292. [PMID: 37714309 DOI: 10.1016/j.nbd.2023.106292] [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: 07/28/2023] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023] Open
Abstract
Chorea-acanthocytosis (ChAc) is an inherited neurodegenerative movement disorder caused by VPS13A gene mutations leading to the absence of protein expression. The striatum is the most affected brain region in ChAc patients. However, the study of the VPS13A function in the brain has been poorly addressed. Here we generated a VPS13A knockdown (KD) model and aimed to elucidate the contribution of VPS13A to synaptic plasticity and neuronal communication in the corticostriatal circuit. First, we infected primary cortical neurons with miR30-shRNA against VPS13A and analyzed its effects on neuronal plasticity. VPS13A-KD neurons showed a higher degree of branching than controls, accompanied by decreased BDNF and PSD-95 levels, indicative of synaptic alterations. We then injected AAV-KD bilaterally in the frontal cortex and two different regions of the striatum of mice and analyzed the effects of VPS13A-KD on animal behavior and synaptic plasticity. VPS13A-KD mice showed modification of the locomotor behavior pattern, with increased exploratory behavior and hyperlocomotion. Corticostriatal dysfunction in VPS13A-KD mice was evidenced by impaired striatal long-term depression (LTD) after stimulation of cortical afferents, which was partially recovered by BDNF administration. VPS13A-KD did not lead to neuronal loss in the cortex or the striatum but induced a decrease in the neuronal release of CX3CL1 and triggered a microglial reaction, especially in the striatum. Notably, CX3CL1 administration partially restored the impaired corticostriatal LTD in VPS13A-KD mice. Our results unveil the involvement of VPS13A in neuronal connectivity modifying BDNF and CX3CL1 release. Moreover, the involvement of VPS13A in synaptic plasticity and motor behavior provides key information to further understand not only ChAc pathophysiology but also other neurological disorders.
Collapse
Affiliation(s)
- Esther García-García
- Dept Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, E-08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), E-08036 Barcelona, Spain; Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), E-08036 Barcelona, Spain.
| | - Alba Ramón-Lainez
- Dept Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, E-08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), E-08036 Barcelona, Spain; Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), E-08036 Barcelona, Spain.
| | - Sara Conde-Berriozabal
- Dept Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, E-08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), E-08036 Barcelona, Spain; Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), E-08036 Barcelona, Spain.
| | - Daniel Del Toro
- Dept Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, E-08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), E-08036 Barcelona, Spain; Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), E-08036 Barcelona, Spain.
| | - Georgia Escaramis
- Dept Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, E-08036 Barcelona, Spain; Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP), Ministerio de Ciencia e Innovación, Madrid, Spain.
| | - Albert Giralt
- Dept Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, E-08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), E-08036 Barcelona, Spain; Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), E-08036 Barcelona, Spain.
| | - Mercè Masana
- Dept Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, E-08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), E-08036 Barcelona, Spain; Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), E-08036 Barcelona, Spain.
| | - Jordi Alberch
- Dept Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, E-08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), E-08036 Barcelona, Spain; Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), E-08036 Barcelona, Spain; Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, E-08036 Barcelona, Spain.
| | - Manuel J Rodríguez
- Dept Biomedical Sciences, School of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, E-08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), E-08036 Barcelona, Spain; Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), E-08036 Barcelona, Spain.
| |
Collapse
|
8
|
Nashiry MA, Sumi SS, Alyami SA, Moni MA. Systems biology approach discovers comorbidity interaction of Parkinson's disease with psychiatric disorders utilizing brain transcriptome. Front Mol Neurosci 2023; 16:1232805. [PMID: 37654790 PMCID: PMC10466791 DOI: 10.3389/fnmol.2023.1232805] [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: 06/01/2023] [Accepted: 07/12/2023] [Indexed: 09/02/2023] Open
Abstract
Several studies found that most patients with Parkinson's disorder (PD) appear to have psychiatric symptoms such as depression, anxiety, hallucination, delusion, and cognitive dysfunction. Therefore, recognizing these psychiatrically symptoms of PD patients is crucial for both symptomatic therapy and better knowledge of the pathophysiology of PD. In order to address this issue, we created a bioinformatics framework to determine the effects of PD mRNA expression on understanding its relationship with psychiatric symptoms in PD patients. We have discovered a significant overlap between the sets of differentially expressed genes from PD exposed tissue and psychiatric disordered tissues using RNA-seq datasets. We have chosen Bipolar disorder and Schizophrenia as psychiatric disorders in our study. A number of significant correlations between PD and the occurrence of psychiatric diseases were also found by gene set enrichment analysis, investigations of the protein-protein interaction network, gene regulatory network, and protein-chemical agent interaction network. We anticipate that the results of this pathogenetic study will provide crucial information for understanding the intricate relationship between PD and psychiatric diseases.
Collapse
Affiliation(s)
- Md Asif Nashiry
- Data Analytics, Northern Alberta Institute of Technology, Edmonton, AB, Canada
| | - Shauli Sarmin Sumi
- Computer Science and Engineering, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Salem A. Alyami
- Mathematics and Statistics, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Mohammad Ali Moni
- Artificial Intelligence and Data Science, Faculty of Health and Behavioural Sciences, School of Health and Rehabilitation Sciences, The University of Queensland, Saint Lucia, QLD, Australia
- Artificial Intelligence and Cyber Futures Institute, Charles Stuart University, Bathurst, NSW, Australia
| |
Collapse
|
9
|
Fan Y, Si Z, Wang L, Zhang L. DYT- TOR1A dystonia: an update on pathogenesis and treatment. Front Neurosci 2023; 17:1216929. [PMID: 37638318 PMCID: PMC10448058 DOI: 10.3389/fnins.2023.1216929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
DYT-TOR1A dystonia is a neurological disorder characterized by involuntary muscle contractions and abnormal movements. It is a severe genetic form of dystonia caused by mutations in the TOR1A gene. TorsinA is a member of the AAA + family of adenosine triphosphatases (ATPases) involved in a variety of cellular functions, including protein folding, lipid metabolism, cytoskeletal organization, and nucleocytoskeletal coupling. Almost all patients with TOR1A-related dystonia harbor the same mutation, an in-frame GAG deletion (ΔGAG) in the last of its 5 exons. This recurrent variant results in the deletion of one of two tandem glutamic acid residues (i.e., E302/303) in a protein named torsinA [torsinA(△E)]. Although the mutation is hereditary, not all carriers will develop DYT-TOR1A dystonia, indicating the involvement of other factors in the disease process. The current understanding of the pathophysiology of DYT-TOR1A dystonia involves multiple factors, including abnormal protein folding, signaling between neurons and glial cells, and dysfunction of the protein quality control system. As there are currently no curative treatments for DYT-TOR1A dystonia, progress in research provides insight into its pathogenesis, leading to potential therapeutic and preventative strategies. This review summarizes the latest research advances in the pathogenesis, diagnosis, and treatment of DYT-TOR1A dystonia.
Collapse
Affiliation(s)
- Yuhang Fan
- Department of Neurology, the Second Hospital of Jilin University, Changchun, China
| | - Zhibo Si
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, China
| | - Linlin Wang
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lei Zhang
- Department of Neurology, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Pocratsky AM, Nascimento F, Özyurt MG, White IJ, Sullivan R, O’Callaghan BJ, Smith CC, Surana S, Beato M, Brownstone RM. Pathophysiology of Dyt1- Tor1a dystonia in mice is mediated by spinal neural circuit dysfunction. Sci Transl Med 2023; 15:eadg3904. [PMID: 37134150 PMCID: PMC7614689 DOI: 10.1126/scitranslmed.adg3904] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/14/2023] [Indexed: 05/05/2023]
Abstract
Dystonia, a neurological disorder defined by abnormal postures and disorganized movements, is considered to be a neural circuit disorder with dysfunction arising within and between multiple brain regions. Given that spinal neural circuits constitute the final pathway for motor control, we sought to determine their contribution to this movement disorder. Focusing on the most common inherited form of dystonia in humans, DYT1-TOR1A, we generated a conditional knockout of the torsin family 1 member A (Tor1a) gene in the mouse spinal cord and dorsal root ganglia (DRG). We found that these mice recapitulated the phenotype of the human condition, developing early-onset generalized torsional dystonia. Motor signs emerged early in the mouse hindlimbs before spreading caudo-rostrally to affect the pelvis, trunk, and forelimbs throughout postnatal maturation. Physiologically, these mice bore the hallmark features of dystonia, including spontaneous contractions at rest and excessive and disorganized contractions, including cocontractions of antagonist muscle groups, during voluntary movements. Spontaneous activity, disorganized motor output, and impaired monosynaptic reflexes, all signs of human dystonia, were recorded from isolated mouse spinal cords from these conditional knockout mice. All components of the monosynaptic reflex arc were affected, including motor neurons. Given that confining the Tor1a conditional knockout to DRG did not lead to early-onset dystonia, we conclude that the pathophysiological substrate of this mouse model of dystonia lies in spinal neural circuits. Together, these data provide new insights into our current understanding of dystonia pathophysiology.
Collapse
Affiliation(s)
- Amanda M. Pocratsky
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London; London, WC1N 3BG, UK
| | - Filipe Nascimento
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London; London, WC1N 3BG, UK
| | - M. Görkem Özyurt
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London; London, WC1N 3BG, UK
| | - Ian J. White
- Laboratory for Molecular Cell Biology, University College London; London, WC1E 6BT, UK
| | - Roisin Sullivan
- Department of Molecular Neuroscience, UCL Queen Square Institute of Neurology, University College London; London, WC1N 3BG, UK
| | - Benjamin J. O’Callaghan
- Department of Molecular Neuroscience, UCL Queen Square Institute of Neurology, University College London; London, WC1N 3BG, UK
| | - Calvin C. Smith
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London; London, WC1N 3BG, UK
| | - Sunaina Surana
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London; London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London; London, WC1E 6BT, UK
| | - Marco Beato
- Department of Neuroscience, Physiology, and Pharmacology, University College London; London, WC1E 6BT, UK
| | - Robert M. Brownstone
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London; London, WC1N 3BG, UK
| |
Collapse
|
11
|
Kim S, Phan S, Shaw TR, Ellisman MH, Veatch SL, Barmada SJ, Pappas SS, Dauer WT. TorsinA is essential for the timing and localization of neuronal nuclear pore complex biogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538491. [PMID: 37162852 PMCID: PMC10168336 DOI: 10.1101/2023.04.26.538491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Nuclear pore complexes (NPCs) regulate information transfer between the nucleus and cytoplasm. NPC defects are linked to several neurological diseases, but the processes governing NPC biogenesis and spatial organization are poorly understood. Here, we identify a temporal window of strongly upregulated NPC biogenesis during neuronal maturation. We demonstrate that the AAA+ protein torsinA, whose loss of function causes the neurodevelopmental movement disorder DYT-TOR1A (DYT1) dystonia, coordinates NPC spatial organization during this period without impacting total NPC density. Using a new mouse line in which endogenous Nup107 is Halo-Tagged, we find that torsinA is essential for correct localization of NPC formation. In the absence of torsinA, the inner nuclear membrane buds excessively at sites of mislocalized, nascent NPCs, and NPC assembly completion is delayed. Our work implies that NPC spatial organization and number are independently regulated and suggests that torsinA is critical for the normal localization and assembly kinetics of NPCs.
Collapse
Affiliation(s)
- Sumin Kim
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Sébastien Phan
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA
| | - Thomas R. Shaw
- Department of Biophysics, University of Michigan, Ann Arbor, MI
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA
| | - Sarah L. Veatch
- Department of Biophysics, University of Michigan, Ann Arbor, MI
| | - Sami J. Barmada
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Samuel S. Pappas
- Peter O’Donnell Jr. Brain Institute, UT Southwestern, Dallas, TX
- Department of Neurology, UT Southwestern, Dallas, TX
| | - William T. Dauer
- Peter O’Donnell Jr. Brain Institute, UT Southwestern, Dallas, TX
- Department of Neurology, UT Southwestern, Dallas, TX
- Department of Neuroscience, UT Southwestern, Dallas, TX
| |
Collapse
|
12
|
Rauschenberger L, Krenig EM, Stengl A, Knorr S, Harder TH, Steeg F, Friedrich MU, Grundmann-Hauser K, Volkmann J, Ip CW. Peripheral nerve injury elicits microstructural and neurochemical changes in the striatum and substantia nigra of a DYT-TOR1A mouse model with dystonia-like movements. Neurobiol Dis 2023; 179:106056. [PMID: 36863527 DOI: 10.1016/j.nbd.2023.106056] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/17/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
The relationship between genotype and phenotype in DYT-TOR1A dystonia as well as the associated motor circuit alterations are still insufficiently understood. DYT-TOR1A dystonia has a remarkably reduced penetrance of 20-30%, which has led to the second-hit hypothesis emphasizing an important role of extragenetic factors in the symptomatogenesis of TOR1A mutation carriers. To analyze whether recovery from a peripheral nerve injury can trigger a dystonic phenotype in asymptomatic hΔGAG3 mice, which overexpress human mutated torsinA, a sciatic nerve crush was applied. An observer-based scoring system as well as an unbiased deep-learning based characterization of the phenotype showed that recovery from a sciatic nerve crush leads to significantly more dystonia-like movements in hΔGAG3 animals compared to wildtype control animals, which persisted over the entire monitored period of 12 weeks. In the basal ganglia, the analysis of medium spiny neurons revealed a significantly reduced number of dendrites, dendrite length and number of spines in the naïve and nerve-crushed hΔGAG3 mice compared to both wildtype control groups indicative of an endophenotypical trait. The volume of striatal calretinin+ interneurons showed alterations in hΔGAG3 mice compared to the wt groups. Nerve-injury related changes were found for striatal ChAT+, parvalbumin+ and nNOS+ interneurons in both genotypes. The dopaminergic neurons of the substantia nigra remained unchanged in number across all groups, however, the cell volume was significantly increased in nerve-crushed hΔGAG3 mice compared to naïve hΔGAG3 mice and wildtype littermates. Moreover, in vivo microdialysis showed an increase of dopamine and its metabolites in the striatum comparing nerve-crushed hΔGAG3 mice to all other groups. The induction of a dystonia-like phenotype in genetically predisposed DYT-TOR1A mice highlights the importance of extragenetic factors in the symptomatogenesis of DYT-TOR1A dystonia. Our experimental approach allowed us to dissect microstructural and neurochemical abnormalities in the basal ganglia, which either reflected a genetic predisposition or endophenotype in DYT-TOR1A mice or a correlate of the induced dystonic phenotype. In particular, neurochemical and morphological changes of the nigrostriatal dopaminergic system were correlated with symptomatogenesis.
Collapse
Affiliation(s)
- Lisa Rauschenberger
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Esther-Marie Krenig
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Alea Stengl
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Susanne Knorr
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Tristan H Harder
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Felix Steeg
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Maximilian U Friedrich
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Kathrin Grundmann-Hauser
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076, Germany; Centre for Rare Diseases, University of Tübingen, 72076, Germany
| | - Jens Volkmann
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany.
| |
Collapse
|
13
|
El Atiallah I, Bonsi P, Tassone A, Martella G, Biella G, Castagno AN, Pisani A, Ponterio G. Synaptic Dysfunction in Dystonia: Update From Experimental Models. Curr Neuropharmacol 2023; 21:2310-2322. [PMID: 37464831 PMCID: PMC10556390 DOI: 10.2174/1570159x21666230718100156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 07/20/2023] Open
Abstract
Dystonia, the third most common movement disorder, refers to a heterogeneous group of neurological diseases characterized by involuntary, sustained or intermittent muscle contractions resulting in repetitive twisting movements and abnormal postures. In the last few years, several studies on animal models helped expand our knowledge of the molecular mechanisms underlying dystonia. These findings have reinforced the notion that the synaptic alterations found mainly in the basal ganglia and cerebellum, including the abnormal neurotransmitters signalling, receptor trafficking and synaptic plasticity, are a common hallmark of different forms of dystonia. In this review, we focus on the major contribution provided by rodent models of DYT-TOR1A, DYT-THAP1, DYT-GNAL, DYT/ PARK-GCH1, DYT/PARK-TH and DYT-SGCE dystonia, which reveal that an abnormal motor network and synaptic dysfunction represent key elements in the pathophysiology of dystonia.
Collapse
Affiliation(s)
- Ilham El Atiallah
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Gerardo Biella
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Antonio N. Castagno
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Fondazione Mondino, Pavia, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Fondazione Mondino, Pavia, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
14
|
Yellajoshyula D, Opeyemi S, Dauer WT, Pappas SS. Genetic evidence of aberrant striatal synaptic maturation and secretory pathway alteration in a dystonia mouse model. DYSTONIA 2022; 1:10892. [PMID: 36874764 PMCID: PMC9980434 DOI: 10.3389/dyst.2022.10892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Animal models of DYT-TOR1A dystonia consistently demonstrate abnormalities of striatal cholinergic function, but the molecular pathways underlying this pathophysiology are unclear. To probe these molecular pathways in a genetic model of DYT-TOR1A, we performed laser microdissection in juvenile mice to isolate striatal cholinergic interneurons and non-cholinergic striatal tissue largely comprising spiny projection neurons during maturation. Both cholinergic and GABAergic enriched samples demonstrated a defined set of gene expression changes consistent with a role of torsinA in the secretory pathway. GABAergic enriched striatum samples also showed alteration to genes regulating synaptic transmission and an upregulation of activity dependent immediate early genes. Reconstruction of Golgi-Cox stained striatal spiny projection neurons from adult mice demonstrated significantly increased spiny density, suggesting that torsinA null striatal neurons have increased excitability during striatal maturation and long lasting increases in afferent input. These findings are consistent with a developmental role for torsinA in the secretory pathway and link torsinA loss of function with functional and structural changes of striatal cholinergic and GABAergic neurons. These transcriptomic datasets are freely available as a resource for future studies of torsinA loss of function-mediated striatal dysfunction.
Collapse
Affiliation(s)
| | - Sunday Opeyemi
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - William T. Dauer
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Samuel S. Pappas
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
15
|
Deep brain stimulation in animal models of dystonia. Neurobiol Dis 2022; 175:105912. [DOI: 10.1016/j.nbd.2022.105912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/19/2022] Open
|
16
|
Al-Wardat M, Schirinzi T, Hadoush H, Kassab M, Yabroudi MA, Opara J, Nawrat-Szołtysik A, Khalil H, Etoom M. Home-Based Exercise to Improve Motor Functions, Cognitive Functions, and Quality of Life in People with Huntington's Disease: A Systematic Review and Meta-Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14915. [PMID: 36429634 PMCID: PMC9690643 DOI: 10.3390/ijerph192214915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 06/16/2023]
Abstract
Exercise in different settings has become a fundamental part of Huntington's disease (HD) management. The aim of this systematic review and meta-analysis was to investigate the effectiveness of home-based exercises (HBE) in HD. Randomized controlled trials (RCTs) investigating the effect of HBE on motor, cognitive, or health-related quality of life (QoL) outcomes in HD were included. Standardized mean difference (SMD), the 95% confidence interval, and p-values were calculated by comparing the outcomes change between HBE and control groups. Seven RCTs met the inclusion criteria. The included RCTs prescribed different types of HBEs, i.e., aerobic strengthening, walking, balance, and fine motor exercises. The HBE protocol length was between 6 and 36 weeks. The meta-analyses showed a significant effect of HBE intervention on motor function measure by Unified Huntington Disease Rating and overall QoL measure by Short Form-36 post-treatment respectively, [SMD = 0.481, p = 0.048], [SMD = 0.378, p = 0.003]. The pooled analysis did not detect significant changes in cognition, gait characteristics, or functional balance scales. The current study shows the positive effect of HBE in HD, especially on motor function and QoL. No significant adverse events were reported. The current results support the clinical effect of HBE intervention on motor function and QoL in HD patients. However, these results should be taken with caution due to the limited available evidence. Well-designed clinical studies that consider the disease severity and stages are required in the future.
Collapse
Affiliation(s)
- Mohammad Al-Wardat
- Department of Rehabilitation Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid P.O. Box 3030, Jordan
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Hikmat Hadoush
- Department of Rehabilitation Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid P.O. Box 3030, Jordan
| | - Manal Kassab
- Department of Maternal and Child Health, Faculty of Nursing, Jordan University of Science and Technology, Irbid P.O. Box 3030, Jordan
| | - Mohammad A. Yabroudi
- Department of Rehabilitation Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid P.O. Box 3030, Jordan
| | - Józef Opara
- Department of Physiotherapy, The Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland
| | | | - Hanan Khalil
- Department of Physical Therapy and Rehabilitation Sciences, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Mohammad Etoom
- Physical Therapy Department, Aqaba University of Technology, Aqaba 77110, Jordan
| |
Collapse
|
17
|
Rodríguez-Urgellés E, Rodríguez-Navarro I, Ballasch I, Del Toro D, Del Castillo I, Brito V, Alberch J, Giralt A. Postnatal Foxp2 regulates early psychiatric-like phenotypes and associated molecular alterations in the R6/1 transgenic mouse model of Huntington's disease. Neurobiol Dis 2022; 173:105854. [PMID: 36029989 DOI: 10.1016/j.nbd.2022.105854] [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: 05/06/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Huntington's Disease (HD) is a devastating disorder characterized by a triad of motor, psychiatric and cognitive manifestations. Psychiatric and emotional symptoms appear at early stages of the disease which are consistently described by patients and caregivers among the most disabling. Here, we show for the first time that Foxp2 is strongly associated with some psychiatric-like disturbances in the R6/1 mouse model of HD. First, 4-week-old (juvenile) R6/1 mice behavioral phenotype was characterized by an increased impulsive-like behavior and less aggressive-like behavior. In this line, we identified an early striatal downregulation of Foxp2 protein starting as soon as at postnatal day 15 that could explain such deficiencies. Interestingly, the rescue of striatal Foxp2 levels from postnatal stages completely reverted the impulsivity-phenotype and partially the social impairments concomitant with a rescue of dendritic spine pathology. A mass spectrometry study indicated that the rescue of spine loss was associated with an improvement of several altered proteins related with cytoskeleton dynamics. Finally, we reproduced and mimicked the impulsivity and social deficits in wild type mice by reducing their striatal Foxp2 expression from postnatal stages. Overall, these results imply that early postnatal reduction of Foxp2 might contribute to the appearance of some of the early psychiatric symptoms in HD.
Collapse
Affiliation(s)
- Ened Rodríguez-Urgellés
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Irene Rodríguez-Navarro
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Iván Ballasch
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Daniel Del Toro
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Ignacio Del Castillo
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Verónica Brito
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain; Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, 08036 Barcelona, Spain.
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain; Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, 08036 Barcelona, Spain.
| |
Collapse
|
18
|
Dzinovic I, Winkelmann J, Zech M. Genetic intersection between dystonia and neurodevelopmental disorders: Insights from genomic sequencing. Parkinsonism Relat Disord 2022; 102:131-140. [DOI: 10.1016/j.parkreldis.2022.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 10/15/2022]
|
19
|
Ponterio G, Faustini G, El Atiallah I, Sciamanna G, Meringolo M, Tassone A, Imbriani P, Cerri S, Martella G, Bonsi P, Bellucci A, Pisani A. Alpha-Synuclein is Involved in DYT1 Dystonia Striatal Synaptic Dysfunction. Mov Disord 2022; 37:949-961. [PMID: 35420219 PMCID: PMC9323501 DOI: 10.1002/mds.29024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/24/2022] [Accepted: 03/19/2022] [Indexed: 12/26/2022] Open
Abstract
Background The neuronal protein alpha‐synuclein (α‐Syn) is crucially involved in Parkinson's disease pathophysiology. Intriguingly, torsinA (TA), the protein causative of DYT1 dystonia, has been found to accumulate in Lewy bodies and to interact with α‐Syn. Both proteins act as molecular chaperones and control synaptic machinery. Despite such evidence, the role of α‐Syn in dystonia has never been investigated. Objective We explored whether α‐Syn and N‐ethylmaleimide sensitive fusion attachment protein receptor proteins (SNAREs), that are known to be modulated by α‐Syn, may be involved in DYT1 dystonia synaptic dysfunction. Methods We used electrophysiological and biochemical techniques to study synaptic alterations in the dorsal striatum of the Tor1a+/Δgag mouse model of DYT1 dystonia. Results In the Tor1a+/Δgag DYT1 mutant mice, we found a significant reduction of α‐Syn levels in whole striata, mainly involving glutamatergic corticostriatal terminals. Strikingly, the striatal levels of the vesicular SNARE VAMP‐2, a direct α‐Syn interactor, and of the transmembrane SNARE synaptosome‐associated protein 23 (SNAP‐23), that promotes glutamate synaptic vesicles release, were markedly decreased in mutant mice. Moreover, we detected an impairment of miniature glutamatergic postsynaptic currents (mEPSCs) recorded from striatal spiny neurons, in parallel with a decreased asynchronous release obtained by measuring quantal EPSCs (qEPSCs), which highlight a robust alteration in release probability. Finally, we also observed a significant reduction of TA striatal expression in α‐Syn null mice. Conclusions Our data demonstrate an unprecedented relationship between TA and α‐Syn, and reveal that α‐Syn and SNAREs alterations characterize the synaptic dysfunction underlying DYT1 dystonia. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.
Collapse
Affiliation(s)
- Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Ilham El Atiallah
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Sciamanna
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy.,UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Imbriani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Pisani
- IRCCS Fondazione Mondino, Pavia, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
20
|
Scarduzio M, Hess EJ, Standaert DG, Eskow Jaunarajs KL. Striatal synaptic dysfunction in dystonia and levodopa-induced dyskinesia. Neurobiol Dis 2022; 166:105650. [DOI: 10.1016/j.nbd.2022.105650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 12/16/2022] Open
|
21
|
Quartarone A, Ghilardi MF. Neuroplasticity in dystonia: Motor symptoms and beyond. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:207-218. [PMID: 35034735 DOI: 10.1016/b978-0-12-819410-2.00031-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This chapter first focuses on the role of altered neuroplasticity mechanisms and their regulation in the genesis of motor symptoms in the various forms of dystonia. In particular, a review of the available literature about focal dystonia suggests that use-dependent plasticity may become detrimental and produce dystonia when practice and repetition are excessive and predisposing conditions are present. Interestingly, recent evidence also shows that functional or psychogenic dystonia, despite the normal plasticity in the sensorimotor system, is characterized by plasticity-related dysfunction within limbic regions. Finally, this chapter reviews the non-motor symptoms that often accompany the motor features of dystonia, including depression and anxiety as well as obsessive-compulsive disorders, pain, and cognitive dysfunctions. Based on the current understanding of these symptoms, we discuss the evidence of their possible relationship to maladaptive plasticity in non-motor basal ganglia circuits involved in their genesis.
Collapse
Affiliation(s)
- Angelo Quartarone
- Department of Biomedical, Dental Sciences and Morphological and Functional Images, University of Messina, Messina, Italy.
| | - Maria Felice Ghilardi
- Department of Molecular, Cellular, and Biomedical Sciences, City University of New York School of Medicine and Neuroscience Program, Graduate Center of the City University of New York, New York, NY, United States
| |
Collapse
|
22
|
Sival DA, Noort SAMV, Tijssen MAJ, de Koning TJ, Verbeek DS. Developmental neurobiology of cerebellar and Basal Ganglia connections. Eur J Paediatr Neurol 2022; 36:123-129. [PMID: 34954622 DOI: 10.1016/j.ejpn.2021.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 10/03/2021] [Accepted: 12/01/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND The high prevalence of mixed phenotypes of Early Onset Ataxia (EOA) with comorbid dystonia has shifted the pathogenetic concept from the cerebellum towards the interconnected cerebellar motor network. This paper on EOA with comorbid dystonia (EOA-dystonia) explores the conceptual relationship between the motor phenotype and the cortico-basal-ganglia-ponto-cerebellar network. METHODS In EOA-dystonia, we reviewed anatomic-, genetic- and biochemical-studies on the comorbidity between ataxia and dystonia. RESULTS In a clinical EOA cohort, the prevalence of dystonia was over 60%. Both human and animal studies converge on the underlying role for the cortico-basal-ganglia-ponto-cerebellar network. Genetic -clinical and -in silico network studies reveal underlying biological pathways for energy production and neural signal transduction. CONCLUSIONS EOA-dystonia phenotypes are attributable to the cortico-basal-ganglia-ponto-cerebellar network, instead of to the cerebellum, alone. The underlying anatomic and pathogenetic pathways have clinical implications for our understanding of the heterogeneous phenotype, neuro-metabolic and genetic testing and potentially also for new treatment strategies, including neuro-modulation.
Collapse
Affiliation(s)
- Deborah A Sival
- Department of Pediatrics, University of Groningen, Groningen, the Netherlands.
| | - Suus A M van Noort
- Department of Neurology and University of Groningen, Groningen, the Netherlands
| | - Marina A J Tijssen
- Department of Neurology and University of Groningen, Groningen, the Netherlands
| | - Tom J de Koning
- Department of Neurology and University of Groningen, Groningen, the Netherlands
| | - Dineke S Verbeek
- Genetics University Medical Center, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
23
|
Sciamanna G, El Atiallah I, Montanari M, Pisani A. Plasticity, genetics and epigenetics in dystonia: An update. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:199-206. [PMID: 35034734 DOI: 10.1016/b978-0-12-819410-2.00011-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dystonia represents a group of movement disorders characterized by involuntary muscle contractions that result in abnormal posture and twisting movements. In the last 20 years several animal models have been generated, greatly improving our knowledge of the neural and molecular mechanism underlying this pathological condition, but the pathophysiology remains still poorly understood. In this review we will discuss recent genetic factors related to dystonia and the current understanding of synaptic plasticity alterations reported by both clinical and experimental research. We will also present recent evidence involving epigenetics mechanisms in dystonia.
Collapse
Affiliation(s)
- Giuseppe Sciamanna
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Ilham El Atiallah
- Department of Systems Medicine, University of Rome 2 Tor Vergata, Rome, Italy
| | - Martina Montanari
- Department of Systems Medicine, University of Rome 2 Tor Vergata, Rome, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Movement Disorders Research Center, IRCCS Mondino Foundation, Pavia, Italy.
| |
Collapse
|
24
|
Rauschenberger L, Knorr S, Pisani A, Hallett M, Volkmann J, Ip CW. Second hit hypothesis in dystonia: Dysfunctional cross talk between neuroplasticity and environment? Neurobiol Dis 2021; 159:105511. [PMID: 34537328 DOI: 10.1016/j.nbd.2021.105511] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 01/08/2023] Open
Abstract
One of the great mysteries in dystonia pathophysiology is the role of environmental factors in disease onset and development. Progress has been made in defining the genetic components of dystonic syndromes, still the mechanisms behind the discrepant relationship between dystonic genotype and phenotype remain largely unclear. Within this review, the preclinical and clinical evidence for environmental stressors as disease modifiers in dystonia pathogenesis are summarized and critically evaluated. The potential role of extragenetic factors is discussed in monogenic as well as adult-onset isolated dystonia. The available clinical evidence for a "second hit" is analyzed in light of the reduced penetrance of monogenic dystonic syndromes and put into context with evidence from animal and cellular models. The contradictory studies on adult-onset dystonia are discussed in detail and backed up by evidence from animal models. Taken together, there is clear evidence of a gene-environment interaction in dystonia, which should be considered in the continued quest to unravel dystonia pathophysiology.
Collapse
Affiliation(s)
- Lisa Rauschenberger
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Susanne Knorr
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Italy; IRCCS Mondino Foundation, Pavia, Italy
| | - Mark Hallett
- Human Motor Control Section, Medical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jens Volkmann
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Straße 11, 97080 Würzburg, Germany.
| |
Collapse
|
25
|
Caffall ZF, Wilkes BJ, Hernández-Martinez R, Rittiner JE, Fox JT, Wan KK, Shipman MK, Titus SA, Zhang YQ, Patnaik S, Hall MD, Boxer MB, Shen M, Li Z, Vaillancourt DE, Calakos N. The HIV protease inhibitor, ritonavir, corrects diverse brain phenotypes across development in mouse model of DYT-TOR1A dystonia. Sci Transl Med 2021; 13:13/607/eabd3904. [PMID: 34408078 DOI: 10.1126/scitranslmed.abd3904] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/14/2020] [Accepted: 06/03/2021] [Indexed: 12/22/2022]
Abstract
Dystonias are a group of chronic movement-disabling disorders for which highly effective oral medications or disease-modifying therapies are lacking. The most effective treatments require invasive procedures such as deep brain stimulation. In this study, we used a high-throughput assay based on a monogenic form of dystonia, DYT1 (DYT-TOR1A), to screen a library of compounds approved for use in humans, the NCATS Pharmaceutical Collection (NPC; 2816 compounds), and identify drugs able to correct mislocalization of the disease-causing protein variant, ∆E302/3 hTorsinA. The HIV protease inhibitor, ritonavir, was among 18 compounds found to normalize hTorsinA mislocalization. Using a DYT1 knock-in mouse model to test efficacy on brain pathologies, we found that ritonavir restored multiple brain abnormalities across development. Ritonavir acutely corrected striatal cholinergic interneuron physiology in the mature brain and yielded sustained correction of diffusion tensor magnetic resonance imaging signals when delivered during a discrete early developmental window. Mechanistically, we found that, across the family of HIV protease inhibitors, efficacy correlated with integrated stress response activation. These preclinical results identify ritonavir as a drug candidate for dystonia with disease-modifying potential.
Collapse
Affiliation(s)
- Zachary F Caffall
- Department of Neurology, Duke University Medical Center, Durham, NC 27715, USA
| | - Bradley J Wilkes
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | | | - Joseph E Rittiner
- Department of Neurology, Duke University Medical Center, Durham, NC 27715, USA
| | - Jennifer T Fox
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Kanny K Wan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Miranda K Shipman
- Department of Neurology, Duke University Medical Center, Durham, NC 27715, USA
| | - Steven A Titus
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Ya-Qin Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Samarjit Patnaik
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Matthew B Boxer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Zhuyin Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - David E Vaillancourt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA.,Department of Neurology, Fixel Institute for Neurological Diseases, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Nicole Calakos
- Department of Neurology, Duke University Medical Center, Durham, NC 27715, USA. .,Department of Neurobiology, Duke University Medical Center, Durham, NC 27715, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27715, USA.,Duke Institute for Brain Sciences, Duke University, Durham, NC 27715, USA
| |
Collapse
|
26
|
Mazere J, Dilharreguy B, Catheline G, Vidailhet M, Deffains M, Vimont D, Ribot B, Barse E, Cif L, Mazoyer B, Langbour N, Pisani A, Allard M, Lamare F, Guehl D, Fernandez P, Burbaud P. Striatal and cerebellar vesicular acetylcholine transporter expression is disrupted in human DYT1 dystonia. Brain 2021; 144:909-923. [PMID: 33638639 DOI: 10.1093/brain/awaa465] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/03/2020] [Accepted: 10/23/2020] [Indexed: 12/20/2022] Open
Abstract
Early-onset torsion dystonia (TOR1A/DYT1) is a devastating hereditary motor disorder whose pathophysiology remains unclear. Studies in transgenic mice suggested abnormal cholinergic transmission in the putamen, but this has not yet been demonstrated in humans. The role of the cerebellum in the pathophysiology of the disease has also been highlighted but the involvement of the intrinsic cerebellar cholinergic system is unknown. In this study, cholinergic neurons were imaged using PET with 18F-fluoroethoxybenzovesamicol, a radioligand of the vesicular acetylcholine transporter (VAChT). Here, we found an age-related decrease in VAChT expression in the posterior putamen and caudate nucleus of DYT1 patients versus matched controls, with low expression in young but not in older patients. In the cerebellar vermis, VAChT expression was also significantly decreased in patients versus controls, but independently of age. Functional connectivity within the motor network studied in MRI and the interregional correlation of VAChT expression studied in PET were also altered in patients. These results show that the cholinergic system is disrupted in the brain of DYT1 patients and is modulated over time through plasticity or compensatory mechanisms.
Collapse
Affiliation(s)
- Joachim Mazere
- Department of Nuclear Medicine, CHU de Bordeaux, France.,Institute of Cognitive and Integrative Neurosciences, CNRS UMR 5287, Bordeaux University, F33000, Bordeaux, France
| | - Bixente Dilharreguy
- Institute of Cognitive and Integrative Neurosciences, CNRS UMR 5287, Bordeaux University, F33000, Bordeaux, France
| | - Gwenaëlle Catheline
- Institute of Cognitive and Integrative Neurosciences, CNRS UMR 5287, Bordeaux University, F33000, Bordeaux, France
| | - Marie Vidailhet
- Institut du Cerveau et de la Moelle épinière (ICM) UMR 1127, hôpital de la Pitié-Salpétrière, Department of Neurology, AP-HP, Sorbonne Université, 75013, Paris, France
| | - Marc Deffains
- Institut des Maladies Neurodégénératives (IMN, CNRS U5393), Université de Bordeaux, 33076, Bordeaux, France
| | - Delphine Vimont
- Department of Nuclear Medicine, CHU de Bordeaux, France.,Institute of Cognitive and Integrative Neurosciences, CNRS UMR 5287, Bordeaux University, F33000, Bordeaux, France
| | - Bastien Ribot
- Institut des Maladies Neurodégénératives (IMN, CNRS U5393), Université de Bordeaux, 33076, Bordeaux, France
| | - Elodie Barse
- Institute of Cognitive and Integrative Neurosciences, CNRS UMR 5287, Bordeaux University, F33000, Bordeaux, France
| | - Laura Cif
- Department of Neurosurgery, CHU de Montpellier, 34000, France
| | - Bernard Mazoyer
- Institut des Maladies Neurodégénératives (IMN, CNRS U5393), Université de Bordeaux, 33076, Bordeaux, France
| | - Nicolas Langbour
- Centre de Recherche en Psychiatrie, CH de la Milétrie, 86000, Poitiers, France
| | - Antonio Pisani
- Department of Brain and Behavioural Sciences, University of Pavia, Italy.,IRCCS Mondino Foundation, Pavia, Italy
| | - Michèle Allard
- Department of Nuclear Medicine, CHU de Bordeaux, France.,Institute of Cognitive and Integrative Neurosciences, CNRS UMR 5287, Bordeaux University, F33000, Bordeaux, France
| | - Frédéric Lamare
- Department of Nuclear Medicine, CHU de Bordeaux, France.,Institute of Cognitive and Integrative Neurosciences, CNRS UMR 5287, Bordeaux University, F33000, Bordeaux, France
| | - Dominique Guehl
- Institut des Maladies Neurodégénératives (IMN, CNRS U5393), Université de Bordeaux, 33076, Bordeaux, France.,Service de Neurophysiologie Clinique, Pôle des Neurosciences Cliniques, CHU de Bordeaux, Bordeaux, France
| | - Philippe Fernandez
- Department of Nuclear Medicine, CHU de Bordeaux, France.,Institute of Cognitive and Integrative Neurosciences, CNRS UMR 5287, Bordeaux University, F33000, Bordeaux, France
| | - Pierre Burbaud
- Institut des Maladies Neurodégénératives (IMN, CNRS U5393), Université de Bordeaux, 33076, Bordeaux, France.,Service de Neurophysiologie Clinique, Pôle des Neurosciences Cliniques, CHU de Bordeaux, Bordeaux, France
| |
Collapse
|
27
|
McClelland VM, Lin JP. Sensorimotor Integration in Childhood Dystonia and Dystonic Cerebral Palsy-A Developmental Perspective. Front Neurol 2021; 12:668081. [PMID: 34367047 PMCID: PMC8343097 DOI: 10.3389/fneur.2021.668081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/07/2021] [Indexed: 11/15/2022] Open
Abstract
Dystonia is a disorder of sensorimotor integration, involving dysfunction within the basal ganglia, cortex, cerebellum, or their inter-connections as part of the sensorimotor network. Some forms of dystonia are also characterized by maladaptive or exaggerated plasticity. Development of the neuronal processes underlying sensorimotor integration is incompletely understood but involves activity-dependent modeling and refining of sensorimotor circuits through processes that are already taking place in utero and which continue through infancy, childhood, and into adolescence. Several genetic dystonias have clinical onset in early childhood, but there is evidence that sensorimotor circuit development may already be disrupted prenatally in these conditions. Dystonic cerebral palsy (DCP) is a form of acquired dystonia with perinatal onset during a period of rapid neurodevelopment and activity-dependent refinement of sensorimotor networks. However, physiological studies of children with dystonia are sparse. This discussion paper addresses the role of neuroplasticity in the development of sensorimotor integration with particular focus on the relevance of these mechanisms for understanding childhood dystonia, DCP, and implications for therapy selection, including neuromodulation and timing of intervention.
Collapse
Affiliation(s)
- Verity M McClelland
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Children's Neurosciences Department, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Jean-Pierre Lin
- Children's Neurosciences Department, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
28
|
Tassone A, Martella G, Meringolo M, Vanni V, Sciamanna G, Ponterio G, Imbriani P, Bonsi P, Pisani A. Vesicular Acetylcholine Transporter Alters Cholinergic Tone and Synaptic Plasticity in DYT1 Dystonia. Mov Disord 2021; 36:2768-2779. [PMID: 34173686 PMCID: PMC9291835 DOI: 10.1002/mds.28698] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 11/17/2022] Open
Abstract
Background Acetylcholine‐mediated transmission plays a central role in the impairment of corticostriatal synaptic activity and plasticity in multiple DYT1 mouse models. However, the nature of such alteration remains unclear. Objective The aim of the present work was to characterize the mechanistic basis of cholinergic dysfunction in DYT1 dystonia to identify potential targets for pharmacological intervention. Methods We utilized electrophysiology recordings, immunohistochemistry, enzymatic activity assays, and Western blotting techniques to analyze in detail the cholinergic machinery in the dorsal striatum of the Tor1a+/− mouse model of DYT1 dystonia. Results We found a significant increase in the vesicular acetylcholine transporter (VAChT) protein level, the protein responsible for loading acetylcholine (ACh) from the cytosol into synaptic vesicles, which indicates an altered cholinergic tone. Accordingly, in Tor1a+/− mice we measured a robust elevation in basal ACh content coupled to a compensatory enhancement of acetylcholinesterase (AChE) enzymatic activity. Moreover, pharmacological activation of dopamine D2 receptors, which is expected to reduce ACh levels, caused an abnormal elevation in its content, as compared to controls. Patch‐clamp recordings revealed a reduced effect of AChE inhibitors on cholinergic interneuron excitability, whereas muscarinic autoreceptor function was preserved. Finally, we tested the hypothesis that blockade of VAChT could restore corticostriatal long‐term synaptic plasticity deficits. Vesamicol, a selective VAChT inhibitor, rescued a normal expression of synaptic plasticity. Conclusions Overall, our findings indicate that VAChT is a key player in the alterations of striatal plasticity and a novel target to normalize cholinergic dysfunction observed in DYT1 dystonia. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Valentina Vanni
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppe Sciamanna
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Imbriani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
29
|
Rescue of striatal long-term depression by chronic mGlu5 receptor negative allosteric modulation in distinct dystonia models. Neuropharmacology 2021; 192:108608. [PMID: 33991565 DOI: 10.1016/j.neuropharm.2021.108608] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/28/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022]
Abstract
An impairment of long-term synaptic plasticity is considered as a peculiar endophenotype of distinct forms of dystonia, a common, disabling movement disorder. Among the few therapeutic options, broad-spectrum antimuscarinic drugs are utilized, aimed at counteracting abnormal striatal acetylcholine-mediated transmission, which plays a crucial role in dystonia pathophysiology. We previously demonstrated a complete loss of long-term synaptic depression (LTD) at corticostriatal synapses in rodent models of two distinct forms of isolated dystonia, resulting from mutations in the TOR1A (DYT1), and GNAL (DYT25) genes. In addition to anticholinergic agents, the aberrant excitability of striatal cholinergic cells can be modulated by group I metabotropic glutamate receptor subtypes (mGlu1 and 5). Here, we tested the efficacy of the negative allosteric modulator (NAM) of metabotropic glutamate 5 (mGlu) receptor, dipraglurant (ADX48621) on striatal LTD. We show that, whereas acute treatment failed to rescue LTD, chronic dipraglurant rescued this form of synaptic plasticity both in DYT1 mice and GNAL rats. Our analysis of the pharmacokinetic profile of dipraglurant revealed a relatively short half-life, which led us to uncover a peculiar time-course of recovery based on the timing from last dipraglurant injection. Indeed, striatal spiny projection neurons (SPNs) recorded within 2 h from last administration showed full expression of synaptic plasticity, whilst the extent of recovery progressively diminished when SPNs were recorded 4-6 h after treatment. Our findings suggest that distinct dystonia genes may share common signaling pathway dysfunction. More importantly, they indicate that dipraglurant might be a potential novel therapeutic agent for this disabling disorder.
Collapse
|
30
|
Li J, Levin DS, Kim AJ, Pappas SS, Dauer WT. TorsinA restoration in a mouse model identifies a critical therapeutic window for DYT1 dystonia. J Clin Invest 2021; 131:139606. [PMID: 33529159 DOI: 10.1172/jci139606] [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] [Received: 11/16/2020] [Accepted: 01/27/2021] [Indexed: 12/18/2022] Open
Abstract
In inherited neurodevelopmental diseases, pathogenic processes unique to critical periods during early brain development may preclude the effectiveness of gene modification therapies applied later in life. We explored this question in a mouse model of DYT1 dystonia, a neurodevelopmental disease caused by a loss-of-function mutation in the TOR1A gene encoding torsinA. To define the temporal requirements for torsinA in normal motor function and gene replacement therapy, we developed a mouse line enabling spatiotemporal control of the endogenous torsinA allele. Suppressing torsinA during embryogenesis caused dystonia-mimicking behavioral and neuropathological phenotypes. Suppressing torsinA during adulthood, however, elicited no discernible abnormalities, establishing an essential requirement for torsinA during a developmental critical period. The developing CNS exhibited a parallel "therapeutic critical period" for torsinA repletion. Although restoring torsinA in juvenile DYT1 mice rescued motor phenotypes, there was no benefit from adult torsinA repletion. These data establish a unique requirement for torsinA in the developing nervous system and demonstrate that the critical period genetic insult provokes permanent pathophysiology mechanistically delinked from torsinA function. These findings imply that to be effective, torsinA-based therapeutic strategies must be employed early in the course of DYT1 dystonia.
Collapse
Affiliation(s)
- Jay Li
- Medical Scientist Training Program.,Cellular and Molecular Biology Graduate Program
| | - Daniel S Levin
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Samuel S Pappas
- Peter O'Donnell Jr. Brain Institute.,Department of Neurology
| | - William T Dauer
- Peter O'Donnell Jr. Brain Institute.,Department of Neurology.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
31
|
Ding B, Tang Y, Ma S, Akter M, Liu ML, Zang T, Zhang CL. Disease Modeling with Human Neurons Reveals LMNB1 Dysregulation Underlying DYT1 Dystonia. J Neurosci 2021; 41:2024-2038. [PMID: 33468570 PMCID: PMC7939088 DOI: 10.1523/jneurosci.2507-20.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 02/08/2023] Open
Abstract
DYT1 dystonia is a hereditary neurologic movement disorder characterized by uncontrollable muscle contractions. It is caused by a heterozygous mutation in Torsin A (TOR1A), a gene encoding a membrane-embedded ATPase. While animal models provide insights into disease mechanisms, significant species-dependent differences exist since animals with the identical heterozygous mutation fail to show pathology. Here, we model DYT1 by using human patient-specific cholinergic motor neurons (MNs) that are generated through either direct conversion of patients' skin fibroblasts or differentiation of induced pluripotent stem cells (iPSCs). These human MNs with the heterozygous TOR1A mutation show reduced neurite length and branches, markedly thickened nuclear lamina, disrupted nuclear morphology, and impaired nucleocytoplasmic transport (NCT) of mRNAs and proteins, whereas they lack the perinuclear "blebs" that are often observed in animal models. Furthermore, we uncover that the nuclear lamina protein LMNB1 is upregulated in DYT1 cells and exhibits abnormal subcellular distribution in a cholinergic MNs-specific manner. Such dysregulation of LMNB1 can be recapitulated by either ectopic expression of the mutant TOR1A gene or shRNA-mediated downregulation of endogenous TOR1A in healthy control MNs. Interestingly, downregulation of LMNB1 can largely ameliorate all the cellular defects in DYT1 MNs. These results reveal the value of disease modeling with human patient-specific neurons and indicate that dysregulation of LMNB1, a crucial component of the nuclear lamina, may constitute a major molecular mechanism underlying DYT1 pathology.SIGNIFICANCE STATEMENT Inaccessibility to patient neurons greatly impedes our understanding of the pathologic mechanisms for dystonia. In this study, we employ reprogrammed human patient-specific motor neurons (MNs) to model DYT1, the most severe hereditary form of dystonia. Our results reveal disease-dependent deficits in nuclear morphology and nucleocytoplasmic transport (NCT). Most importantly, we further identify LMNB1 dysregulation as a major contributor to these deficits, uncovering a new pathologic mechanism for DYT1 dystonia.
Collapse
Affiliation(s)
- Baojin Ding
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana 70503
| | - Yu Tang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Shuaipeng Ma
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Masuma Akter
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana 70503
| | - Meng-Lu Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Tong Zang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Chun-Li Zhang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
32
|
Li J, Kim S, Pappas SS, Dauer WT. CNS critical periods: implications for dystonia and other neurodevelopmental disorders. JCI Insight 2021; 6:142483. [PMID: 33616084 PMCID: PMC7934928 DOI: 10.1172/jci.insight.142483] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Critical periods are discrete developmental stages when the nervous system is especially sensitive to stimuli that facilitate circuit maturation. The distinctive landscapes assumed by the developing CNS create analogous periods of susceptibility to pathogenic insults and responsiveness to therapy. Here, we review critical periods in nervous system development and disease, with an emphasis on the neurodevelopmental disorder DYT1 dystonia. We highlight clinical and laboratory observations supporting the existence of a critical period during which the DYT1 mutation is uniquely harmful, and the implications for future therapeutic development.
Collapse
Affiliation(s)
- Jay Li
- Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Sumin Kim
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | | | - William T. Dauer
- Peter O’Donnell Jr. Brain Institute
- Department of Neurology, and
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
33
|
Ferrazzoli D, Ortelli P, Volpe D, Cucca A, Versace V, Nardone R, Saltuari L, Sebastianelli L. The Ties That Bind: Aberrant Plasticity and Networks Dysfunction in Movement Disorders-Implications for Rehabilitation. Brain Connect 2021; 11:278-296. [PMID: 33403893 DOI: 10.1089/brain.2020.0971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: Movement disorders encompass various conditions affecting the nervous system. The pathological processes underlying movement disorders lead to aberrant synaptic plastic changes, which in turn alter the functioning of large-scale brain networks. Therefore, clinical phenomenology does not only entail motor symptoms but also cognitive and motivational disturbances. The result is the disruption of motor learning and motor behavior. Due to this complexity, the responsiveness to standard therapies could be disappointing. Specific forms of rehabilitation entailing goal-based practice, aerobic training, and the use of noninvasive brain stimulation techniques could "restore" neuroplasticity at motor-cognitive circuitries, leading to clinical gains. This is probably associated with modulations occurring at both molecular (synaptic) and circuitry levels (networks). Several gaps remain in our understanding of the relationships among plasticity and neural networks and how neurorehabilitation could promote clinical gains is still unclear. Purposes: In this review, we outline first the networks involved in motor learning and behavior and analyze which mechanisms link the pathological synaptic plastic changes with these networks' disruption in movement disorders. Therefore, we provide theoretical and practical bases to be applied for treatment in rehabilitation.
Collapse
Affiliation(s)
- Davide Ferrazzoli
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Vipiteno-Sterzing, Italy
| | - Paola Ortelli
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Vipiteno-Sterzing, Italy
| | - Daniele Volpe
- Fresco Parkinson Center, Villa Margherita, S. Stefano Riabilitazione, Vicenza, Italy
| | - Alberto Cucca
- Fresco Parkinson Center, Villa Margherita, S. Stefano Riabilitazione, Vicenza, Italy.,Department of Neurology, The Marlene & Paolo Fresco Institute for Parkinson's & Movement Disorders, NYU School of Medicine, New York, New York, USA.,Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Viviana Versace
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Vipiteno-Sterzing, Italy
| | - Raffaele Nardone
- Department of Neurology, Franz Tappeiner Hospital (SABES-ASDAA), Merano-Meran, Italy.,Department of Neurology, Christian Doppler Medical Center, Paracelsus University Salzburg, Salzburg, Austria
| | - Leopold Saltuari
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Vipiteno-Sterzing, Italy
| | - Luca Sebastianelli
- Department of Neurorehabilitation, Hospital of Vipiteno (SABES-ASDAA), Vipiteno-Sterzing, Italy
| |
Collapse
|
34
|
Baumann H, Ott F, Weber J, Trilck-Winkler M, Münchau A, Zittel S, Kostić VS, Kaiser FJ, Klein C, Busch H, Seibler P, Lohmann K. Linking Penetrance and Transcription in DYT-THAP1: Insights From a Human iPSC-Derived Cortical Model. Mov Disord 2021; 36:1381-1391. [PMID: 33547842 DOI: 10.1002/mds.28506] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/20/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The THAP1 gene encodes a transcription factor, and pathogenic variants cause a form of autosomal dominant, isolated dystonia (DYT-THAP1) with reduced penetrance. Factors underlying both reduced penetrance and the disease mechanism of DYT-THAP1 are largely unknown. METHODS We performed transcriptome analysis on 29 cortical neuronal precursors derived from human-induced pluripotent stem cell lines generated from manifesting and nonmanifesting THAP1 mutation carriers and control individuals. RESULTS Whole transcriptome analysis showed a penetrance-linked signature with expressional changes more pronounced in the group of manifesting (MMCs) than in nonmanifesting mutation carriers (NMCs) when compared to controls. A direct comparison of the transcriptomes in MMCs versus NMCs showed significant upregulation of the DRD4 gene in MMCs. A gene set enrichment analysis demonstrated alterations in various neurotransmitter release cycle pathways, extracellular matrix organization, and deoxyribonucleic acid methylation between MMCs and NMCs. When specifically considering transcription factors, the expression of YY1 and SIX2 differed in MMCs versus NMCs. Further, THAP1 was upregulated in the group of MMCs. CONCLUSIONS To our knowledge, this is the first report systematically analyzing reduced penetrance in DYT-THAP1 in a human model using transcriptomes. Our findings indicate that transcriptional alterations during cortical development influence DYT-THAP1 pathogenesis and penetrance. We reinforce previously linked pathways including dopamine and eukaryotic translation initiation factor 2 alpha signaling in the pathogenesis of dystonia including DYT-THAP1 and suggest extracellular matrix organization and deoxyribonucleic acid methylation as mediators of disease protection. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Hauke Baumann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Fabian Ott
- Institute of Experimental Dermatology and Institute of Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Joachim Weber
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | | | - Alexander Münchau
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.,Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Simone Zittel
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Frank J Kaiser
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Institute of Human Genetics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Hauke Busch
- Institute of Experimental Dermatology and Institute of Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
35
|
Liu Y, Xing H, Yokoi F, Vaillancourt DE, Li Y. Investigating the role of striatal dopamine receptor 2 in motor coordination and balance: Insights into the pathogenesis of DYT1 dystonia. Behav Brain Res 2021; 403:113137. [PMID: 33476687 DOI: 10.1016/j.bbr.2021.113137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/29/2020] [Accepted: 01/13/2021] [Indexed: 12/16/2022]
Abstract
DYT1 or DYT-TOR1A dystonia is early-onset, generalized dystonia. Most DYT1 dystonia patients have a heterozygous trinucleotide GAG deletion in DYT1 or TOR1A gene, with a loss of a glutamic acid residue of the protein torsinA. DYT1 dystonia patients show reduced striatal dopamine D2 receptor (D2R) binding activity. We previously reported reduced striatal D2R proteins and impaired corticostriatal plasticity in Dyt1 ΔGAG heterozygous knock-in (Dyt1 KI) mice. It remains unclear how the D2R reduction contributes to the pathogenesis of DYT1 dystonia. Recent knockout studies indicate that D2R on cholinergic interneurons (Chls) has a significant role in corticostriatal plasticity, while D2R on medium spiny neurons (MSNs) plays a minor role. To determine how reduced D2Rs on ChIs and MSNs affect motor performance, we generated ChI- or MSN-specific D2R conditional knockout mice (Drd2 ChKO or Drd2 sKO). The striatal ChIs in the Drd2 ChKO mice showed an increased firing frequency and impaired quinpirole-induced inhibition, suggesting a reduced D2R function on the ChIs. Drd2 ChKO mice had an age-dependent deficient performance on the beam-walking test similar to the Dyt1 KI mice. The Drd2 sKO mice, conversely, had a deficit on the rotarod but not the beam-walking test. Our findings suggest that D2Rs on Chls and MSNs have critical roles in motor control and balance. The similarity of the beam-walking deficit between the Drd2 ChKO and Dyt1 KI mice supports our earlier notion that D2R reduction on striatal ChIs contributes to the pathophysiology and the motor symptoms of DYT1 dystonia.
Collapse
Affiliation(s)
- Yuning Liu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States; Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Hong Xing
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Fumiaki Yokoi
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - David E Vaillancourt
- Department of Applied Physiology and Kinesiology, Biomedical Engineering, and Neurology, University of Florida, Gainesville, FL, United States
| | - Yuqing Li
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, United States; Genetics Institute, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
36
|
Melis C, Beauvais G, Muntean BS, Cirnaru MD, Otrimski G, Creus-Muncunill J, Martemyanov KA, Gonzalez-Alegre P, Ehrlich ME. Striatal Dopamine Induced ERK Phosphorylation Is Altered in Mouse Models of Monogenic Dystonia. Mov Disord 2021; 36:1147-1157. [PMID: 33458877 DOI: 10.1002/mds.28476] [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] [Received: 10/16/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Similar to some monogenic forms of dystonia, levodopa-induced dyskinesia is a hyperkinetic movement disorder with abnormal nigrostriatal dopaminergic neurotransmission. Molecularly, it is characterized by hyper-induction of phosphorylation of extracellular signal-related kinase in response to dopamine in medium spiny neurons of the direct pathway. OBJECTIVES The objective of this study was to determine if mouse models of monogenic dystonia exhibit molecular features of levodopa-induced dyskinesia. METHODS Western blotting and quantitative immunofluorescence was used to assay baseline and/or dopamine-induced levels of the phosphorylated kinase in the striatum in mouse models of DYT1, DYT6, and DYT25 expressing a reporter in dopamine D1 receptor-expressing projection neurons. Cyclic adenosine monophosphate (cAMP) immunoassay and adenylyl cyclase activity assays were also performed. RESULTS In DYT1 and DYT6 models, blocking dopamine reuptake with cocaine leads to enhanced extracellular signal-related kinase phosphorylation in dorsomedial striatal medium spiny neurons in the direct pathway, which is abolished by pretreatment with the N-methyl-d-aspartate antagonist MK-801. Phosphorylation is decreased in a model of DYT25. Levels of basal and stimulated cAMP and adenylyl cyclase activity were normal in the DYT1 and DYT6 mice and decreased in the DYT25 mice. Oxotremorine induced increased abnormal movements in the DYT1 knock-in mice. CONCLUSIONS The increased dopamine induction of extracellular signal-related kinase phosphorylation in 2 genetic types of dystonia, similar to what occurs in levodopa-induced dyskinesia, and its decrease in a third, suggests that abnormal signal transduction in response to dopamine in the postsynaptic nigrostriatal pathway might be a point of convergence for dystonia and other hyperkinetic movement disorders, potentially offering common therapeutic targets. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chiara Melis
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Genevieve Beauvais
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Garrett Otrimski
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Pedro Gonzalez-Alegre
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
37
|
Effect of Gabapentin in a Neuropathic Pain Model in Mice Overexpressing Human Wild-Type or Human Mutated Torsin A. Life (Basel) 2021; 11:life11010041. [PMID: 33445430 PMCID: PMC7826569 DOI: 10.3390/life11010041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND DYT1 dystonia is the most common form of early-onset inherited dystonia, which is caused by mutation of torsin A (TA) belonging to the "ATPases associated with a variety of cellular activities" (AAA + ATPase). Dystonia is often accompanied by pain, and neuropathic pain can be associated to peripherally induced movement disorder and dystonia. However, no evidence exists on the effect of gabapentin in mice subjected to neuropathic pain model overexpressing human normal or mutated TA. METHODS Mice subjected to L5 spinal nerve ligation (SNL) develop mechanical allodynia and upregulation of the α2δ-1 L-type calcium channel subunit, forming a validated experimental model of neuropathic pain. Under these experimental conditions, TA is expressed in dorsal horn neurons and astrocytes and colocalizes with α2δ-1. Similar to this subunit, TA is overexpressed in dorsal horn 7 days after SNL. This model has been used to investigate (1) basal mechanical sensitivity; (2) neuropathic pain phases; and (3) the effect of gabapentin, an α2δ-1 ligand used against neuropathic pain, in non-transgenic (NT) C57BL/6 mice and in mice overexpressing human wild-type (hWT) or mutant (hMT) TA. RESULTS In comparison to non-transgenic mice, the threshold for mechanical sensitivity in hWT or hMT does not differ (Kruskal-Wallis test = 1.478; p = 0.4777, although, in the latter animals, neuropathic pain recovery phase is delayed. Interestingly, gabapentin (100 mg/Kg) reduces allodynia at its peak (occurring between post-operative day 7 and day 10) but not in the phase of recovery. CONCLUSIONS These data lend support to the investigation on the role of TA in the molecular machinery engaged during neuropathic pain.
Collapse
|
38
|
Induction of BDNF Expression in Layer II/III and Layer V Neurons of the Motor Cortex Is Essential for Motor Learning. J Neurosci 2020; 40:6289-6308. [PMID: 32651187 PMCID: PMC7424868 DOI: 10.1523/jneurosci.0288-20.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022] Open
Abstract
Motor learning depends on synaptic plasticity between corticostriatal projections and striatal medium spiny neurons. Retrograde tracing from the dorsolateral striatum reveals that both layer II/III and V neurons in the motor cortex express BDNF as a potential regulator of plasticity in corticostriatal projections in male and female mice. The number of these BDNF-expressing cortical neurons and levels of BDNF protein are highest in juvenile mice when adult motor patterns are shaped, while BDNF levels in the adult are low. When mice are trained by physical exercise in the adult, BDNF expression in motor cortex is reinduced, especially in layer II/III projection neurons. Reduced expression of cortical BDNF in 3-month-old mice results in impaired motor learning while space memory is preserved. These findings suggest that activity regulates BDNF expression differentially in layers II/III and V striatal afferents from motor cortex and that cortical BDNF is essential for motor learning. SIGNIFICANCE STATEMENT Motor learning in mice depends on corticostriatal BDNF supply, and regulation of BDNF expression during motor learning is highest in corticostriatal projection neurons in cortical layer II/III.
Collapse
|
39
|
Perl S, Richter F, Richter A. Striatal and cortical metabotropic glutamate 5 receptor expression and behavioral effects of the positive allosteric modulator CDPPB in a model of DYT1 dystonia. Pharmacol Biochem Behav 2020; 196:172977. [PMID: 32615137 DOI: 10.1016/j.pbb.2020.172977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/09/2020] [Accepted: 06/23/2020] [Indexed: 11/19/2022]
Abstract
The metabotropic glutamate 5 (mGlu5) receptor is critically involved in corticostriatal plasticity which is disturbed in various animal models of dystonia. Recently, the positive allosteric modulator 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) exerted prodyskinetic effects in a phenotypic model of episodic dystonia. In the DYT1 knock-in (KI) mouse, a model for a persistent type of dystonia, previous ex vivo electrophysiological experiments indicated that mGlu5 receptors are involved in abnormal striatal plasticity. Therefore, in the present study we examined the mGlu5 receptor expression in the striatum and cortex of DYT1 KI mice in comparison with wildtype littermates. By immunohistochemistry (IHC) we found a lower expression of mGlu5 receptors in the cortex (16%) and ventral striatum (10%) but not in the whole striatum of DYT1 KI mice, while mRNA levels were merely lower in the striatum of DYT1 KI mice (43%). However, mGlu5 receptor protein levels measured by western blotting showed no significant differences in tissue of the whole striatum and in the cortex between both genotypes. Since DYT1 KI mice do not exhibit dystonic symptoms, we investigated if CDPPB provokes dystonia or dyskinesia. CDPPB (10, 20 and 30 mg/kg intraperitoneal, i.p.) did not induce abnormal movements and the locomotor activity did not differ between DYT1 KI and wildtype mice. The present data do not provide evidence for a crucial role of the mGlu5 receptor in the pathophysiology of DYT1 dystonia, but corticostriatal changes are in line with the hypothesis of maladaptive plasticity in dystonia.
Collapse
Affiliation(s)
- Stefanie Perl
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany.
| | - Franziska Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany; Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
40
|
Mutant Allele-Specific CRISPR Disruption in DYT1 Dystonia Fibroblasts Restores Cell Function. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:1-12. [PMID: 32502938 PMCID: PMC7270506 DOI: 10.1016/j.omtn.2020.05.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/15/2020] [Accepted: 05/12/2020] [Indexed: 12/16/2022]
Abstract
Most individuals affected with DYT1 dystonia have a heterozygous 3-bp deletion in the TOR1A gene (c.907_909delGAG). The mutation appears to act through a dominant-negative mechanism compromising normal torsinA function, and it is proposed that reducing mutant torsinA may normalize torsinA activity. In this study, we used an engineered Cas9 variant from Streptococcus pyogenes (SpCas9-VRQR) to target the mutation in the TOR1A gene in order to disrupt mutant torsinA in DYT1 patient fibroblasts. Selective targeting of the DYT1 allele was highly efficient with most common non-homologous end joining (NHEJ) edits, leading to a predicted premature stop codon with loss of the torsinA C terminus (delta 302–332 aa). Structural analysis predicted a functionally inactive status of this truncated torsinA due to the loss of residues associated with ATPase activity and binding to LULL1. Immunoblotting showed a reduction of the torsinA protein level in Cas9-edited DYT1 fibroblasts, and a functional assay using HSV infection indicated a phenotypic recovery toward that observed in control fibroblasts. These findings suggest that the selective disruption of the mutant TOR1A allele using CRISPR-Cas9 inactivates mutant torsinA, allowing the remaining wild-type torsinA to exert normal function.
Collapse
|
41
|
Li J, Liang CC, Pappas SS, Dauer WT. TorsinB overexpression prevents abnormal twisting in DYT1 dystonia mouse models. eLife 2020; 9:e54285. [PMID: 32202496 PMCID: PMC7141835 DOI: 10.7554/elife.54285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/23/2020] [Indexed: 12/13/2022] Open
Abstract
Genetic redundancy can be exploited to identify therapeutic targets for inherited disorders. We explored this possibility in DYT1 dystonia, a neurodevelopmental movement disorder caused by a loss-of-function (LOF) mutation in the TOR1A gene encoding torsinA. Prior work demonstrates that torsinA and its paralog torsinB have conserved functions at the nuclear envelope. This work established that low neuronal levels of torsinB dictate the neuronal selective phenotype of nuclear membrane budding. Here, we examined whether torsinB expression levels impact the onset or severity of abnormal movements or neuropathological features in DYT1 mouse models. We demonstrate that torsinB levels bidirectionally regulate these phenotypes. Reducing torsinB levels causes a dose-dependent worsening whereas torsinB overexpression rescues torsinA LOF-mediated abnormal movements and neurodegeneration. These findings identify torsinB as a potent modifier of torsinA LOF phenotypes and suggest that augmentation of torsinB expression may retard or prevent symptom development in DYT1 dystonia.
Collapse
Affiliation(s)
- Jay Li
- Medical Scientist Training Program, University of MichiganAnn ArborUnited States
- Cellular and Molecular Biology Graduate Program, University of MichiganAnn ArborUnited States
| | - Chun-Chi Liang
- Department of Neurology, University of MichiganAnn ArborUnited States
| | - Samuel S Pappas
- Peter O’Donnell Jr. Brain Institute, Departments of Neuroscience and Neurology & Neurotherapeutics, University of Texas SouthwesternDallasUnited States
| | - William T Dauer
- Department of Neurology, University of MichiganAnn ArborUnited States
- Peter O’Donnell Jr. Brain Institute, Departments of Neuroscience and Neurology & Neurotherapeutics, University of Texas SouthwesternDallasUnited States
| |
Collapse
|
42
|
Synaptic GluN2A-Containing NMDA Receptors: From Physiology to Pathological Synaptic Plasticity. Int J Mol Sci 2020; 21:ijms21041538. [PMID: 32102377 PMCID: PMC7073220 DOI: 10.3390/ijms21041538] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
N-Methyl-d-Aspartate Receptors (NMDARs) are ionotropic glutamate-gated receptors. NMDARs are tetramers composed by several homologous subunits of GluN1-, GluN2-, or GluN3-type, leading to the existence in the central nervous system of a high variety of receptor subtypes with different pharmacological and signaling properties. NMDAR subunit composition is strictly regulated during development and by activity-dependent synaptic plasticity. Given the differences between GluN2 regulatory subunits of NMDAR in several functions, here we will focus on the synaptic pool of NMDARs containing the GluN2A subunit, addressing its role in both physiology and pathological synaptic plasticity as well as the contribution in these events of different types of GluN2A-interacting proteins.
Collapse
|
43
|
Rauschenberger L, Knorr S, Al-Zuraiqi Y, Tovote P, Volkmann J, Ip CW. Striatal dopaminergic dysregulation and dystonia-like movements induced by sensorimotor stress in a pharmacological mouse model of rapid-onset dystonia-parkinsonism. Exp Neurol 2020; 323:113109. [DOI: 10.1016/j.expneurol.2019.113109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/18/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022]
|
44
|
Calabresi P, Standaert DG. Dystonia and levodopa-induced dyskinesias in Parkinson's disease: Is there a connection? Neurobiol Dis 2019; 132:104579. [PMID: 31445160 PMCID: PMC6834901 DOI: 10.1016/j.nbd.2019.104579] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 08/01/2019] [Accepted: 08/14/2019] [Indexed: 11/24/2022] Open
Abstract
Dystonia and levodopa-induced dyskinesia (LID) are both hyperkinetic movement disorders. Dystonia arises most often spontaneously, although it may be seen after stroke, injury, or as a result of genetic causes. LID is associated with Parkinson's disease (PD), emerging as a consequence of chronic therapy with levodopa, and may be either dystonic or choreiform. LID and dystonia share important phenomenological properties and mechanisms. Both LID and dystonia are generated by an integrated circuit involving the cortex, basal ganglia, thalamus and cerebellum. They also share dysregulation of striatal cholinergic signaling and abnormalities of striatal synaptic plasticity. The long duration nature of both LID and dystonia suggests that there may be underlying epigenetic dysregulation as a proximate cause. While both may improve after interventions such as deep brain stimulation (DBS), neither currently has a satisfactory medical therapy, and many people are disabled by the symptoms of dystonia and LID. Further study of the fundamental mechanisms connecting these two disorders may lead to novel approaches to treatment or prevention.
Collapse
Affiliation(s)
- Paolo Calabresi
- Neurological Clinic, Department of Medicine, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia 06132, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
45
|
Yu-Taeger L, Ott T, Bonsi P, Tomczak C, Wassouf Z, Martella G, Sciamanna G, Imbriani P, Ponterio G, Tassone A, Schulze-Hentrich JM, Goodchild R, Riess O, Pisani A, Grundmann-Hauser K, Nguyen HP. Impaired dopamine- and adenosine-mediated signaling and plasticity in a novel rodent model for DYT25 dystonia. Neurobiol Dis 2019; 134:104634. [PMID: 31678405 DOI: 10.1016/j.nbd.2019.104634] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/19/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Dystonia is a neurological movement disorder characterized by sustained or intermittent involuntary muscle contractions. Loss-of-function mutations in the GNAL gene have been identified to be the cause of "isolated" dystonia DYT25. The GNAL gene encodes for the guanine nucleotide-binding protein G(olf) subunit alpha (Gαolf), which is mainly expressed in the olfactory bulb and the striatum and functions as a modulator during neurotransmission coupling with D1R and A2AR. Previously, heterozygous Gαolf -deficient mice (Gnal+/-) have been generated and showed a mild phenotype at basal condition. In contrast, homozygous deletion of Gnal in mice (Gnal-/-) resulted in a significantly reduced survival rate. In this study, using the CRISPR-Cas9 system we generated and characterized heterozygous Gnal knockout rats (Gnal+/-) with a 13 base pair deletion in the first exon of the rat Gnal splicing variant 2, a major isoform in both human and rat striatum. Gnal+/- rats showed early-onset phenotypes associated with impaired dopamine transmission, including reduction in locomotor activity, deficits in rotarod performance and an abnormal motor skill learning ability. At cellular and molecular level, we found down-regulated Arc expression, increased cell surface distribution of AMPA receptors, and the loss of D2R-dependent corticostriatal long-term depression (LTD) in Gnal+/- rats. Based on the evidence that D2R activity is normally inhibited by adenosine A2ARs, co-localized on the same population of striatal neurons, we show that blockade of A2ARs restores physiological LTD. This animal model may be a valuable tool for investigating Gαolf function and finding a suitable treatment for dystonia associated with deficient dopamine transmission.
Collapse
Affiliation(s)
- Libo Yu-Taeger
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany; Centre for Rare Diseases (ZSE), University of Tuebingen, Tuebingen, Germany
| | - Thomas Ott
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany; Core Facility Transgenic Animals, University Clinics Tuebingen, Tuebingen, Germany
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Celina Tomczak
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany; Centre for Rare Diseases (ZSE), University of Tuebingen, Tuebingen, Germany
| | - Zinah Wassouf
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany; Centre for Rare Diseases (ZSE), University of Tuebingen, Tuebingen, Germany
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata,Rome, Italy
| | - Giuseppe Sciamanna
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata,Rome, Italy
| | - Paola Imbriani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata,Rome, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata,Rome, Italy
| | - Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata,Rome, Italy
| | - Julia M Schulze-Hentrich
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany; Centre for Rare Diseases (ZSE), University of Tuebingen, Tuebingen, Germany
| | - Rose Goodchild
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; KU Leuven, Dept. Neurosciences, Leuven, Belgium
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany; Centre for Rare Diseases (ZSE), University of Tuebingen, Tuebingen, Germany
| | - Antonio Pisani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata,Rome, Italy
| | - Kathrin Grundmann-Hauser
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany; Centre for Rare Diseases (ZSE), University of Tuebingen, Tuebingen, Germany
| | - Huu Phuc Nguyen
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany; Department of Human Genetics, Faculty of Medicine, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
46
|
A Novel Transgenic Mouse Model to Investigate the Cell-Autonomous Effects of torsinA(ΔE) Expression in Striatal Output Neurons. Neuroscience 2019; 422:1-11. [PMID: 31669362 DOI: 10.1016/j.neuroscience.2019.09.007] [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: 05/24/2019] [Revised: 09/04/2019] [Accepted: 09/10/2019] [Indexed: 11/22/2022]
Abstract
Dystonia is a disabling neurological syndrome characterized by abnormal movements and postures that result from intermittent or sustained involuntary muscle contractions; mutations of DYT1/TOR1A are the most common cause of childhood-onset, generalized, inherited dystonia. Patient and mouse model data strongly support dysregulation of the nigrostriatal dopamine neurotransmission circuit in the presence of the DYT1-causing mutation. To determine striatal medium spiny neuron (MSN) cell-autonomous and non-cell autonomous effects relevant to dopamine transmission, we created a transgenic mouse in which expression of mutant torsinA in forebrain is restricted to MSNs. We assayed electrically evoked and cocaine-enhanced dopamine release and locomotor activity, dopamine uptake, gene expression of dopamine-associated neuropeptides and receptors, and response to the muscarinic cholinergic antagonist, trihexyphenidyl. We found that over-expression of mutant torsinA in MSNs produces complex cell-autonomous and non-cell autonomous alterations in nigrostriatal dopaminergic and intrastriatal cholinergic function, similar to that found in pan-cellular DYT1 mouse models. These data introduce targets for future studies to identify which are causative and which are compensatory in DYT1 dystonia, and thereby aid in defining appropriate therapies.
Collapse
|
47
|
GPRIN3 Controls Neuronal Excitability, Morphology, and Striatal-Dependent Behaviors in the Indirect Pathway of the Striatum. J Neurosci 2019; 39:7513-7528. [PMID: 31363062 DOI: 10.1523/jneurosci.2454-18.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
The regulation of the striatum by the GPCR signaling through neuromodulators is essential for its physiology and physiopathology, so it is necessary to know all the compounds of these pathways. In this study, we identified a new important partner of the dopaminergic pathway: GPRIN3 (a member of the GPRIN family). GPRIN3 is highly expressed in the striatum but with undefined function. Cell sorting of medium spiny neurons (MSNs) in indirect MSNs and direct MSNs indicated the presence of the GPRIN3 gene in both populations with a preferential expression in indirect MSNs. This led us to generate GPRIN3 KO mice by CRISPR/Cas9 and test male animals to access possible alterations in morphological, electrophysiological, and behavioral parameters following its absence. 3D reconstruction analysis of MSNs revealed increased neuronal arborization in GPRIN3 KO and modified passive and active electrophysiological properties. These cellular alterations were coupled with increased motivation and cocaine-induced hyperlocomotion. Additionally, using a specific indirect MSN knockdown, we showed a preferential role for GPRIN3 in indirect MSNs related to the D2R signaling. Together, these results show that GPRIN3 is a mediator of D2R function in the striatum playing a major role in striatal physiology.SIGNIFICANCE STATEMENT The striatum is the main input of the basal ganglia processing information from different brain regions through the combined actions of direct pathway neurons and indirect pathway neurons. Both neuronal populations are defined by the expression of dopamine D1R or D2R GPCRs, respectively. How these neurons signal to the respective G-protein is still debatable. Here we identified GPRIN3 as a putative selective controller of D2R function in the striatum playing a critical role in striatal-associated behaviors and cellular functions. This study represents the identification of a new target to tackle striatal dysfunction associated with the D2R, such as schizophrenia, Parkinson's disease, and drug addiction.
Collapse
|
48
|
The neurobiological basis for novel experimental therapeutics in dystonia. Neurobiol Dis 2019; 130:104526. [PMID: 31279827 DOI: 10.1016/j.nbd.2019.104526] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/13/2019] [Accepted: 07/03/2019] [Indexed: 12/17/2022] Open
Abstract
Dystonia is a movement disorder characterized by involuntary muscle contractions, twisting movements, and abnormal postures that may affect one or multiple body regions. Dystonia is the third most common movement disorder after Parkinson's disease and essential tremor. Despite its relative frequency, small molecule therapeutics for dystonia are limited. Development of new therapeutics is further hampered by the heterogeneity of both clinical symptoms and etiologies in dystonia. Recent advances in both animal and cell-based models have helped clarify divergent etiologies in dystonia and have facilitated the identification of new therapeutic targets. Advances in medicinal chemistry have also made available novel compounds for testing in biochemical, physiological, and behavioral models of dystonia. Here, we briefly review motor circuit anatomy and the anatomical and functional abnormalities in dystonia. We then discuss recently identified therapeutic targets in dystonia based on recent preclinical animal studies and clinical trials investigating novel therapeutics.
Collapse
|
49
|
Gonzalez-Alegre P. Advances in molecular and cell biology of dystonia: Focus on torsinA. Neurobiol Dis 2019; 127:233-241. [DOI: 10.1016/j.nbd.2019.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/20/2019] [Accepted: 03/09/2019] [Indexed: 12/15/2022] Open
|
50
|
Gangarossa G, Perez S, Dembitskaya Y, Prokin I, Berry H, Venance L. BDNF Controls Bidirectional Endocannabinoid Plasticity at Corticostriatal Synapses. Cereb Cortex 2019; 30:197-214. [DOI: 10.1093/cercor/bhz081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022] Open
Abstract
AbstractThe dorsal striatum exhibits bidirectional corticostriatal synaptic plasticity, NMDAR and endocannabinoids (eCB) mediated, necessary for the encoding of procedural learning. Therefore, characterizing factors controlling corticostriatal plasticity is of crucial importance. Brain-derived neurotrophic factor (BDNF) and its receptor, the tropomyosine receptor kinase-B (TrkB), shape striatal functions, and their dysfunction deeply affects basal ganglia. BDNF/TrkB signaling controls NMDAR plasticity in various brain structures including the striatum. However, despite cross-talk between BDNF and eCBs, the role of BDNF in eCB plasticity remains unknown. Here, we show that BDNF/TrkB signaling promotes eCB-plasticity (LTD and LTP) induced by rate-based (low-frequency stimulation) or spike-timing–based (spike-timing–dependent plasticity, STDP) paradigm in striatum. We show that TrkB activation is required for the expression and the scaling of both eCB-LTD and eCB-LTP. Using 2-photon imaging of dendritic spines combined with patch-clamp recordings, we show that TrkB activation prolongs intracellular calcium transients, thus increasing eCB synthesis and release. We provide a mathematical model for the dynamics of the signaling pathways involved in corticostriatal plasticity. Finally, we show that TrkB activation enlarges the domain of expression of eCB-STDP. Our results reveal a novel role for BDNF/TrkB signaling in governing eCB-plasticity expression in striatum and thus the engram of procedural learning.
Collapse
Affiliation(s)
- Giuseppe Gangarossa
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Sylvie Perez
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Yulia Dembitskaya
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Ilya Prokin
- INRIA, Villeurbanne, France
- University of Lyon, LIRIS UMR, Villeurbanne, France
| | - Hugues Berry
- INRIA, Villeurbanne, France
- University of Lyon, LIRIS UMR, Villeurbanne, France
| | - Laurent Venance
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| |
Collapse
|