1
|
Zhu L, Meng H, Zhang W, Xie W, Sun H, Hou S. The pathogenesis of blepharospasm. Front Neurol 2024; 14:1336348. [PMID: 38274886 PMCID: PMC10808626 DOI: 10.3389/fneur.2023.1336348] [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: 11/13/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Blepharospasm is a focal dystonia characterized by involuntary tetanic contractions of the orbicularis oculi muscle, which can lead to functional blindness and loss of independent living ability in severe cases. It usually occurs in adults, with a higher incidence rate in women than in men. The etiology and pathogenesis of this disease have not been elucidated to date, but it is traditionally believed to be related to the basal ganglia. Studies have also shown that this is related to the decreased activity of inhibitory neurons in the cerebral cortex caused by environmental factors and genetic predisposition. Increasingly, studies have focused on the imbalance in the regulation of neurotransmitters, including dopamine, serotonin, and acetylcholine, in blepharospasm. The onset of the disease is insidious, and the misdiagnosis rate is high based on history and clinical manifestations. This article reviews the etiology, epidemiological features, and pathogenesis of blepharospasm, to improve understanding of the disease by neurologists and ophthalmologists.
Collapse
Affiliation(s)
- Lixia Zhu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Hongmei Meng
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Wuqiong Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Wenjing Xie
- Department of Neurology, The Second Hospital of Jilin University, Changchun, China
| | - Huaiyu Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Shuai Hou
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
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: 0.5] [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
|
3
|
Srinivasan SR. Targeting Circuit Abnormalities in Neurodegenerative Disease. Mol Pharmacol 2023; 103:38-44. [PMID: 36310030 DOI: 10.1124/molpharm.122.000563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 02/03/2023] Open
Abstract
Despite significant improvement in our ability to diagnose both common and rare neurodegenerative diseases and understand their underlying biologic mechanisms, there remains a disproportionate lack of effective treatments, reflecting the complexity of these disorders. Successfully advancing novel treatments for neurodegenerative disorders will require reconsideration of traditional approaches, which to date have focused largely on specific disease proteins or cells of origin. This article proposes reframing these diseases as conditions of dysfunctional circuitry as a complement to ongoing efforts. Specifically reviewed is how aberrant spiking is a common downstream mechanism in numerous neurodegenerative diseases, often driven by dysfunction in specific ion channels. Surgical modification of this electrical activity via deep brain stimulation is already an approved modality for many of these disorders. Therefore, restoring proper electrical activity by targeting these channels pharmacologically represents a viable strategy for intervention, not only for symptomatic management but also as a potential disease-modifying therapy. Such an approach is likely to be a promising route to treating these devastating disorders, either as monotherapy or in conjunction with current drugs. SIGNIFICANCE STATEMENT: Despite extensive research and improved understanding of the biology driving neurodegenerative disease, there has not been a concomitant increase in approved therapies. Accordingly, it is time to shift our perspective and recognize these diseases also as disorders of circuitry to further yield novel drug targets and new interventions. An approach focused on treating dysfunctional circuitry has the potential to reduce or reverse patient symptoms and potentially modify disease course.
Collapse
|
4
|
Huang X, Zhang M, Li B, Shang H, Yang J. Structural and functional brain abnormalities in idiopathic cervical dystonia: A multimodal meta-analysis. Parkinsonism Relat Disord 2022; 103:153-165. [DOI: 10.1016/j.parkreldis.2022.08.029] [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: 04/27/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/15/2022]
|
5
|
Cai H, Ni L, Hu X, Ding X. Inhibition of endoplasmic reticulum stress reverses synaptic plasticity deficits in striatum of DYT1 dystonia mice. Aging (Albany NY) 2021; 13:20319-20334. [PMID: 34398825 PMCID: PMC8436893 DOI: 10.18632/aging.203413] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/21/2021] [Indexed: 11/25/2022]
Abstract
Background and objective: Striatal plasticity alterations caused by endoplasmic reticulum (ER) stress is supposed to be critically involved in the mechanism of DYT1 dystonia. In the current study, we expanded this research field by investigating the critical role of ER stress underlying synaptic plasticity impairment imposed by mutant heterozygous Tor1a+/- in a DYT1 dystonia mouse model. Methods: Heterozygous Tor1a+/- mouse model for DYT1 dystonia was established. Wild-type (Tor1a+/+, N=10) and mutant (Tor1a+/-, N=10) mice from post-natal day P25 to P35 were randomly distributed to experimental and control groups. Patch-clamp and current-clamp recordings of SPNs were conducted with intracellular electrodes for electrophysiological analyses. Striatal changes of the direct and indirect pathways were investigated via immunofluorescence. Golgi-Cox staining was conducted to observe spine morphology of SPNs. To quantify postsynaptic signaling proteins in striatum, RNA-Seq, qRT-PCR and WB were performed in striatal tissues. Results: Long-term depression (LTD) was failed to be induced, while long-term potentiation (LTP) was further strengthened in striatal spiny projection neurons (SPNs) from the Tor1a+/- DYT1 dystonia mice. Spine morphology analyses revealed a significant increase of both number of mushroom type spines and spine width in Tor1a+/- SPNs. In addition, increased AMPA receptor function and the reduction of NMDA/AMPA ratio in the postsynaptic of Tor1a+/- SPNs was observed, along with increased ER stress protein levels in striatum of Tor1a+/- DYT1 dystonia mice. Notably, ER stress inhibitors, tauroursodeoxycholic acid (TUDCA), could rescue LTD as well as AMPA currents. Conclusion: The current study illustrated the role of ER stress in mediating structural and functional plasticity alterations in Tor1a+/- SPNs. Inhibition of the ER stress by TUDCA is beneficial in reversing the deficits at the cellular and molecular levels. Remedy of dystonia associated neurological and motor functional impairment by ER stress inhibitors could be a recommendable therapeutic agent in clinical practice.
Collapse
Affiliation(s)
- Huaying Cai
- Department of Neurology, Neuroscience Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Linhui Ni
- Department of Neurology, Neuroscience Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Xingyue Hu
- Department of Neurology, Neuroscience Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Xianjun Ding
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
6
|
Ma H, Qu J, Ye L, Shu Y, Qu Q. Blepharospasm, Oromandibular Dystonia, and Meige Syndrome: Clinical and Genetic Update. Front Neurol 2021; 12:630221. [PMID: 33854473 PMCID: PMC8039296 DOI: 10.3389/fneur.2021.630221] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Meige syndrome (MS) is cranial dystonia characterized by the combination of upper and lower cranial involvement and including binocular eyelid spasms (blepharospasm; BSP) and involuntary movements of the jaw muscles (oromandibular dystonia; OMD). The etiology and pathogenesis of this disorder of the extrapyramidal system are not well-understood. Neurologic and ophthalmic examinations often reveal no abnormalities, making diagnosis difficult and often resulting in misdiagnosis. A small proportion of patients have a family history of the disease, but to date no causative genes have been identified to date and no cure is available, although botulinum toxin A therapy effectively mitigates the symptoms and deep brain stimulation is gaining increasing attention as a viable alternative treatment option. Here we review the history and progress of research on MS, BSP, and OMD, as well as the etiology, pathology, diagnosis, and treatment.
Collapse
Affiliation(s)
- Hongying Ma
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Institute for Rational and Safe Medication Practices, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Liangjun Ye
- Department of Pharmacy, Hunan Provincial Corps Hospital of Chinese People's Armed Police Force, Changsha, China
| | - Yi Shu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Institute for Rational and Safe Medication Practices, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
7
|
Jinnah H, Sun YV. Dystonia genes and their biological pathways. Neurobiol Dis 2019; 129:159-168. [DOI: 10.1016/j.nbd.2019.05.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/05/2019] [Accepted: 05/17/2019] [Indexed: 12/27/2022] Open
|
8
|
Khan MM, Xiao J, Hollingsworth TJ, Patel D, Selley DE, Ring TL, LeDoux MS. Gnal haploinsufficiency causes genomic instability and increased sensitivity to haloperidol. Exp Neurol 2019; 318:61-70. [PMID: 31034808 DOI: 10.1016/j.expneurol.2019.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 04/09/2019] [Accepted: 04/23/2019] [Indexed: 12/26/2022]
Abstract
GNAL encodes guanine nucleotide-binding protein subunit Gα(olf) which plays a key role in striatal medium spiny neuron (MSN)-dopamine signaling. GNAL loss-of-function mutations are causally-associated with isolated dystonia, a movement disorder characterized by involuntary muscle contractions leading to abnormal postures. Dopamine D2 receptor (D2R) blockers such as haloperidol are mainstays in the treatment of psychosis but may contribute to the development of secondary acute and tardive dystonia. Administration of haloperidol promotes cAMP-dependent signaling in D2R-expressing indirect pathway MSNs. At present, little is known about the cellular relationships among isolated, acute, and tardive dystonia. Herein, we report the effects of acute D2R blockade on motor behavior, DNA repair, cAMP-mediated histone H3 phosphorylation (Ser10), and cell death in Gnal+/- mice and their isogenic Gnal+/+ littermates. In comparison to Gnal+/+ littermates, Gnal+/- mice exhibited increased catalepsy responses, persistent DNA breaks, decreased cAMP-dependent histone H3 phosphorylation (Ser10), and increased cell death in response to haloperidol. In striatum, aged Gnal+/- mice exhibited increased global DNA methylation, increased euchromatin, and dendritic structural abnormalities. Our results provide evidence that Gα(olf) deficiency intensifies the effects of D2R antagonism and suggests that loss-of-function variants in GNAL may increase risk for movement disorders associated with D2R blockers. We hypothesize that the effects of Gα(olf) dysfunction and/or long-term D2R antagonism may lead to epigenetic silencing, transcriptional dysregulation, and, ultimately, cellular senescence and/or apoptosis in human brain.
Collapse
Affiliation(s)
- Mohammad Moshahid Khan
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Division of Rehabilitation Sciences, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Jianfeng Xiao
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - T J Hollingsworth
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Damini Patel
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Dana E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Trevor L Ring
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Mark S LeDoux
- Department of Psychology, University of Memphis, Memphis, TN 38152, USA.
| |
Collapse
|
9
|
Khan MM, Xiao J, Patel D, LeDoux MS. DNA damage and neurodegenerative phenotypes in aged Ciz1 null mice. Neurobiol Aging 2018; 62:180-190. [PMID: 29154038 DOI: 10.1016/j.neurobiolaging.2017.10.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/17/2017] [Accepted: 10/16/2017] [Indexed: 12/28/2022]
Abstract
Cell-cycle dysfunction and faulty DNA repair are closely intertwined pathobiological processes that may contribute to several neurodegenerative disorders. CDKN1A interacting zinc finger protein 1 (CIZ1) plays a critical role in DNA replication and cell-cycle progression at the G1/S checkpoint. Germline or somatic variants in CIZ1 have been linked to several neural and extra-neural diseases. Recently, we showed that germline knockout of Ciz1 is associated with motor and hematological abnormalities in young adult mice. However, the effects of CIZ1 deficiency in much older mice may be more relevant to understanding age-related declines in cognitive and motor functioning and age-related neurologic disorders such as isolated dystonia and Alzheimer disease. Mouse embryonic fibroblasts from Ciz1-/- mice showed abnormal sensitivity to the effects of γ-irradiation with persistent DNA breaks, aberrant cell-cycle progression, and apoptosis. Aged (18-month-old) Ciz1-/- mice exhibited marked deficits in motor and cognitive functioning, and, in brain tissues, overt DNA damage, NF-κB upregulation, oxidative stress, vascular dysfunction, inflammation, and cell death. These findings indicate that the deleterious effects of CIZ1 deficiency become more pronounced with aging and suggest that defects of cell-cycle control and associated DNA repair pathways in postmitotic neurons could contribute to global neurologic decline in elderly human populations. Accordingly, the G1/S cell-cycle checkpoint and associated DNA repair pathways may be targets for the prevention and treatment of age-related neurodegenerative processes.
Collapse
Affiliation(s)
- Mohammad Moshahid Khan
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jianfeng Xiao
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Damini Patel
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mark S LeDoux
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
10
|
Yu-Taeger L, Gaiser V, Lotzer L, Roenisch T, Fabry BT, Stricker-Shaver J, Casadei N, Walter M, Schaller M, Riess O, Nguyen HP, Ott T, Grundmann-Hauser K. Dynamic nuclear envelope phenotype in rats overexpressing mutated human torsinA protein. Biol Open 2018; 7:bio.032839. [PMID: 29739751 PMCID: PMC6078351 DOI: 10.1242/bio.032839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A three-base-pair deletion in the human TOR1A gene is causative for the most common form of primary dystonia: the early-onset dystonia type 1 (DYT1 dystonia). The pathophysiological consequences of this mutation are still unknown. To study the pathology of the mutant torsinA (TOR1A) protein, we have generated a transgenic rat line that overexpresses the human mutant protein under the control of the human TOR1A promoter. This new animal model was phenotyped with several approaches, including behavioral tests and neuropathological analyses. Motor phenotype, cellular and ultrastructural key features of torsinA pathology were found in this new transgenic rat line, supporting that it can be used as a model system for investigating the disease’s development. Analyses of mutant TOR1A protein expression in various brain regions also showed a dynamic expression pattern and a reversible nuclear envelope pathology. These findings suggest the differential vulnerabilities of distinct neuronal subpopulations. Furthermore, the reversibility of the nuclear envelope pathology might be a therapeutic target to treat the disease. Summary: A novel transgenic rat model displaying dystonia-like phenotypes and dynamic processes in NE pathology can become a useful tool for therapy development for dystonia and other related diseases.
Collapse
Affiliation(s)
- Libo Yu-Taeger
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Viktoria Gaiser
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Larissa Lotzer
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Tina Roenisch
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Core Facility Transgenic Animals, University Hospital Tuebingen, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Benedikt Timo Fabry
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Janice Stricker-Shaver
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Nicolas Casadei
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Michael Walter
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Agilent Technologies, 5301 Stevens Creek Blvd, Santa Clara, CA 95051, USA
| | - Martin Schaller
- Department of Dermatology, University of Tuebingen, Liebermeisterstr. 25, 72076 Tuebingen, Germany
| | - Olaf Riess
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Huu Phuc Nguyen
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany .,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| | - Thomas Ott
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Core Facility Transgenic Animals, University Hospital Tuebingen, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Kathrin Grundmann-Hauser
- Institute for Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany.,Centre for Rare Diseases, University of Tuebingen, Calwerstr. 7, 72076 Tuebingen, Germany
| |
Collapse
|
11
|
Maltese M, Stanic J, Tassone A, Sciamanna G, Ponterio G, Vanni V, Martella G, Imbriani P, Bonsi P, Mercuri NB, Gardoni F, Pisani A. Early structural and functional plasticity alterations in a susceptibility period of DYT1 dystonia mouse striatum. eLife 2018; 7:33331. [PMID: 29504938 PMCID: PMC5849413 DOI: 10.7554/elife.33331] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/02/2018] [Indexed: 12/30/2022] Open
Abstract
The onset of abnormal movements in DYT1 dystonia is between childhood and adolescence, although it is unclear why clinical manifestations appear during this developmental period. Plasticity at corticostriatal synapses is critically involved in motor memory. In the Tor1a+/Δgag DYT1 dystonia mouse model, long-term potentiation (LTP) appeared prematurely in a critical developmental window in striatal spiny neurons (SPNs), while long-term depression (LTD) was never recorded. Analysis of dendritic spines showed an increase of both spine width and mature mushroom spines in Tor1a+/Δgag neurons, paralleled by an enhanced AMPA receptor (AMPAR) accumulation. BDNF regulates AMPAR expression during development. Accordingly, both proBDNF and BDNF levels were significantly higher in Tor1a+/Δgag mice. Consistently, antagonism of BDNF rescued synaptic plasticity deficits and AMPA currents. Our findings demonstrate that early loss of functional and structural synaptic homeostasis represents a unique endophenotypic trait during striatal maturation, promoting the appearance of clinical manifestations in mutation carriers.
Collapse
Affiliation(s)
- Marta Maltese
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Jennifer Stanic
- Department of Pharmacology, University of Milan, Milan, Italy
| | - Annalisa Tassone
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppe Sciamanna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giulia Ponterio
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Valentina Vanni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppina Martella
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Imbriani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | - Antonio Pisani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
12
|
Jinnah HA, Hess EJ. Evolving concepts in the pathogenesis of dystonia. Parkinsonism Relat Disord 2018; 46 Suppl 1:S62-S65. [PMID: 28784298 PMCID: PMC5696051 DOI: 10.1016/j.parkreldis.2017.08.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 01/01/2023]
Abstract
INTRODUCTION The dystonias are a group of disorders defined by over-contraction of muscles leading to abnormal movements and postures. In recent years, enormous advances have been made in elucidating the neurobiological mechanisms responsible for many types of dystonia. METHODS A literature review was conducted focusing on evolving concepts in dystonia genetics, anatomy and physiology. RESULTS The list of genes related to dystonia has grown from a relatively small number to more than 100. Concepts regarding the neuroanatomical basis for dystonia have evolved from a relatively narrow focus on dysfunction of the basal ganglia to a broader motor network model in which the basal ganglia, cerebellum, cerebral cortex, and other brain regions play a key role. Physiologically, our understanding of the core abnormalities has matured; and numerous changes in neural signaling have been revealed in the basal ganglia, cerebellum and cortex. CONCLUSION Although the dystonias share certain clinical aspects such as over-contraction of muscles leading to abnormal movements and postures, they actually comprise a very clinically and etiologically heterogeneous group of disorders. Understanding their neurobiological basis is important for devising rational therapies appropriately targeted for specific subgroups of patients.
Collapse
Affiliation(s)
- H A Jinnah
- Departments of Neurology, Human Genetics and Pediatrics, Emory University, Atlanta, GA, USA.
| | - Ellen J Hess
- Department of Pharmacology and Neurology, Emory University, Atlanta, GA, USA
| |
Collapse
|
13
|
Jinnah HA, Neychev V, Hess EJ. The Anatomical Basis for Dystonia: The Motor Network Model. Tremor Other Hyperkinet Mov (N Y) 2017; 7:506. [PMID: 29123945 PMCID: PMC5673689 DOI: 10.7916/d8v69x3s] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/25/2017] [Indexed: 01/27/2023] Open
Abstract
Background The dystonias include a clinically and etiologically very diverse group of disorders. There are both degenerative and non-degenerative subtypes resulting from genetic or acquired causes. Traditionally, all dystonias have been viewed as disorders of the basal ganglia. However, there has been increasing appreciation for involvement of other brain regions including the cerebellum, thalamus, midbrain, and cortex. Much of the early evidence for these other brain regions has come from studies of animals, but multiple recent studies have been done with humans, in an effort to confirm or refute involvement of these other regions. The purpose of this article is to review the new evidence from animals and humans regarding the motor network model, and to address the issues important to translational neuroscience. Methods The English literature was reviewed for articles relating to the neuroanatomical basis for various types of dystonia in both animals and humans. Results There is evidence from both animals and humans that multiple brain regions play an important role in various types of dystonia. The most direct evidence for specific brain regions comes from animal studies using pharmacological, lesion, or genetic methods. In these studies, experimental manipulations of specific brain regions provide direct evidence for involvement of the basal ganglia, cerebellum, thalamus and other regions. Additional evidence also comes from human studies using neuropathological, neuroimaging, non-invasive brain stimulation, and surgical interventions. In these studies, the evidence is less conclusive, because discriminating the regions that cause dystonia from those that reflect secondary responses to abnormal movements is more challenging. Discussion Overall, the evidence from both animals and humans suggests that different regions may play important roles in different subtypes of dystonia. The evidence so far provides strong support for the motor network model. There are obvious challenges, but also advantages, of attempting to translate knowledge gained from animals into a more complete understanding of human dystonia and novel therapeutic strategies.
Collapse
Affiliation(s)
- H. A. Jinnah
- Departments of Neurology, Human Genetics and Pediatrics, Emory University, Atlanta, GA, USA
| | - Vladimir Neychev
- Department of Surgery, University Multiprofile Hospital for Active Treatment “Alexandrovska”, Medical University of Sofia, Sofia, Bulgaria
| | - Ellen J. Hess
- Departments of Pharmacology and Neurology, Emory University, Atlanta, GA, USA
| |
Collapse
|
14
|
Abstract
INTRODUCTION Dystonia is a clinically heterogeneous group of hyperkinetic movement disorders. Recent advances have provided a better understanding of these conditions with significant clinical impact. SOURCES OF DATA Peer reviewed journals and reviews. PubMed.gov. AREAS OF AGREEMENT A recent consensus classification, including the assessment of phenomenology and identification of the dystonia syndromes, has provided a helpful tool for the clinical assessment. New forms of monogenic dystonia have been recently identified. AREAS OF CONTROVERSY Despite recent advances in the understanding of dystonia, treatment remains symptomatic in most patients. GROWING POINTS Recent advances in genetics have provided a better understanding of the potential pathogenic mechanisms involved in dystonia. Deep brain stimulation has shown to improve focal and combined forms of dystonia and its indications are constantly expanding. AREAS TIMELY FOR DEVELOPING RESEARCH Growing understanding of the disease mechanisms involved will allow the development of targeted and disease-modifying therapies in the future.
Collapse
Affiliation(s)
- Eduardo De Pablo-Fernandez
- Reta Lila Weston Institute of Neurological Studies, UCL institute of Neurology, 1 Wakefield Street, WC1N 1PJ London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Institute of Neurology, 1 Wakefield Street, WC1N 1PJ London, UK
| | - Thomas T Warner
- Reta Lila Weston Institute of Neurological Studies, UCL institute of Neurology, 1 Wakefield Street, WC1N 1PJ London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Institute of Neurology, 1 Wakefield Street, WC1N 1PJ London, UK
| |
Collapse
|
15
|
Filip P, Gallea C, Lehéricy S, Bertasi E, Popa T, Mareček R, Lungu OV, Kašpárek T, Vaníček J, Bareš M. Disruption in cerebellar and basal ganglia networks during a visuospatial task in cervical dystonia. Mov Disord 2017; 32:757-768. [DOI: 10.1002/mds.26930] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/06/2016] [Accepted: 12/09/2016] [Indexed: 12/19/2022] Open
Affiliation(s)
- Pavel Filip
- Central European Institute of Technology; Central European Institute of Technology, Masaryk University (CEITEC MU), Behavioral and Social Neuroscience Research Group, Masaryk University; Brno Czech Republic
- First Department of Neurology; Faculty of Medicine, Masaryk University and St. Anne's Teaching Hospital; Brno Czech Republic
| | - Cécile Gallea
- Institut du Cerveau et de la Moelle épinière-ICM, Centre de NeuroImagerie de Recherche-Centre de Neuro-Imagerie de Recherche, Sorbonne Universités, University Pierre and Marie CURIE Univ Paris 06, University of Minnesota Rochester (UMR) S 1127, Centre national de la recherche scientifique (CNRS) UMR 7225, ICM, F-75013, ICM team Control of Normal and Abnormal Movement; Paris France
| | - Stéphane Lehéricy
- Institut du Cerveau et de la Moelle épinière-ICM, Centre de NeuroImagerie de Recherche-Centre de Neuro-Imagerie de Recherche, Sorbonne Universités, University Pierre and Marie CURIE Univ Paris 06, University of Minnesota Rochester (UMR) S 1127, Centre national de la recherche scientifique (CNRS) UMR 7225, ICM, F-75013, ICM team Control of Normal and Abnormal Movement; Paris France
| | - Eric Bertasi
- Institut du Cerveau et de la Moelle épinière-ICM, Centre de NeuroImagerie de Recherche-Centre de Neuro-Imagerie de Recherche, Sorbonne Universités, University Pierre and Marie CURIE Univ Paris 06, University of Minnesota Rochester (UMR) S 1127, Centre national de la recherche scientifique (CNRS) UMR 7225, ICM, F-75013, ICM team Control of Normal and Abnormal Movement; Paris France
| | - Traian Popa
- Institut du Cerveau et de la Moelle épinière-ICM, Centre de NeuroImagerie de Recherche-Centre de Neuro-Imagerie de Recherche, Sorbonne Universités, University Pierre and Marie CURIE Univ Paris 06, University of Minnesota Rochester (UMR) S 1127, Centre national de la recherche scientifique (CNRS) UMR 7225, ICM, F-75013, ICM team Control of Normal and Abnormal Movement; Paris France
| | - Radek Mareček
- Central European Institute of Technology; CEITEC MU, Multimodal and Functional Neuroimaging Research Group, Masaryk University; Brno Czech Republic
| | - Ovidiu V. Lungu
- Department of Psychiatry; Université de Montréal; Montréal Québec Canada
- Functional Neuroimaging Unit; Research Center of the Geriatric Institute affiliated with the Université de Montréal; Montréal Québec Canada
| | - Tomáš Kašpárek
- Central European Institute of Technology; Central European Institute of Technology, Masaryk University (CEITEC MU), Behavioral and Social Neuroscience Research Group, Masaryk University; Brno Czech Republic
- Department of Psychiatry; Faculty of Medicine, Masaryk University and Teaching Hospital Brno; Brno Czech Republic
| | - Jiří Vaníček
- Department of Imaging Methods; Masaryk University and St. Anne's Teaching Hospital; Brno Czech Republic
| | - Martin Bareš
- Central European Institute of Technology; Central European Institute of Technology, Masaryk University (CEITEC MU), Behavioral and Social Neuroscience Research Group, Masaryk University; Brno Czech Republic
- First Department of Neurology; Faculty of Medicine, Masaryk University and St. Anne's Teaching Hospital; Brno Czech Republic
- Department of Neurology; School of Medicine, University of Minnesota; Minneapolis USA
| |
Collapse
|
16
|
Xiao J, Vemula SR, Xue Y, Khan MM, Carlisle FA, Waite AJ, Blake DJ, Dragatsis I, Zhao Y, LeDoux MS. Role of major and brain-specific Sgce isoforms in the pathogenesis of myoclonus-dystonia syndrome. Neurobiol Dis 2016; 98:52-65. [PMID: 27890709 DOI: 10.1016/j.nbd.2016.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 11/06/2016] [Accepted: 11/17/2016] [Indexed: 01/09/2023] Open
Abstract
Loss-of-function mutations in SGCE, which encodes ε-sarcoglycan (ε-SG), cause myoclonus-dystonia syndrome (OMIM159900, DYT11). A "major" ε-SG protein derived from CCDS5637.1 (NM_003919.2) and a "brain-specific" protein, that includes sequence derived from alternative exon 11b (CCDS47642.1, NM_001099400.1), are reportedly localized in post- and pre-synaptic membrane fractions, respectively. Moreover, deficiency of the "brain-specific" isoform and other isoforms derived from exon 11b may be central to the pathogenesis of DYT11. However, no animal model supports this hypothesis. Gene-trapped ES cells (CMHD-GT_148G1-3, intron 9 of NM_011360) were used to generate a novel Sgce mouse model (C57BL/6J background) with markedly reduced expression of isoforms derived from exons 3' to exon 9 of NM_011360. Among those brain regions analyzed in adult (2month-old) wild-type (WT) mice, cerebellum showed the highest relative expression of isoforms incorporating exon 11b. Homozygotes (SgceGt(148G1)Cmhd/Gt(148G1)Cmhd or SgceGt/Gt) and paternal heterozygotes (Sgcem+/pGt, m-maternal, p-paternal) showed 60 to 70% reductions in expression of total Sgce. Although expression of the major (NM_011360) and brain-specific (NM_001130189) isoforms was markedly reduced, expression of short isoforms was preserved and relatively small amounts of chimeric ε-SG/β-galactosidase fusion protein was produced by the Sgce gene-trap locus. Immunoaffinity purification followed by mass spectrometry assessments of Sgcem+/pGt mouse brain using pan- or brain-specific ε-SG antibodies revealed significant reductions of ε-SG and other interacting sarcoglycans. Genome-wide gene-expression data using RNA derived from adult Sgcem+/pGt mouse cerebellum showed that the top up-regulated genes were involved in cell cycle, cellular development, cell death and survival, while the top down-regulated genes were associated with protein synthesis, cellular development, and cell death and survival. In comparison to WT littermates, Sgcem+/pGt mice exhibited "tiptoe" gait and stimulus-induced appendicular posturing between Postnatal Days 14 to 16. Abnormalities noted in older Sgcem+/pGt mice included reduced body weight, altered gait dynamics, and reduced open-field activity. Overt spontaneous or stimulus-sensitive myoclonus was not apparent on the C57BL/6J background or mixed C57BL/6J-BALB/c and C57BL/6J-129S2 backgrounds. Our data confirm that mouse Sgce is a maternally imprinted gene and suggests that short Sgce isoforms may compensate, in part, for deficiency of major and brain-specific Sgce isoforms.
Collapse
Affiliation(s)
- Jianfeng Xiao
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Satya R Vemula
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yi Xue
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Mohammad M Khan
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Francesca A Carlisle
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cathays, Cardiff, CF24 4HQ, Great Britain, United Kingdom
| | - Adrian J Waite
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cathays, Cardiff, CF24 4HQ, Great Britain, United Kingdom
| | - Derek J Blake
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cathays, Cardiff, CF24 4HQ, Great Britain, United Kingdom
| | - Ioannis Dragatsis
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yu Zhao
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Mark S LeDoux
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
17
|
Xiao J, Vemula SR, Xue Y, Khan MM, Kuruvilla KP, Marquez-Lona EM, Cobb MR, LeDoux MS. Motor phenotypes and molecular networks associated with germline deficiency of Ciz1. Exp Neurol 2016; 283:110-20. [PMID: 27163549 DOI: 10.1016/j.expneurol.2016.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 04/20/2016] [Accepted: 05/03/2016] [Indexed: 12/26/2022]
Abstract
A missense mutation in CIZ1 (c.790A>G, p.S264G) was linked to autosomal dominant cervical dystonia in a large multiplex Caucasian pedigree (OMIM614860, DYT23). CIZ1 is a p21((Cip1/Waf1)) -interacting zinc finger protein, widely expressed in neural and extra-neural tissues, and plays a role in DNA synthesis at the G1/S cell-cycle checkpoint. The role of CIZ1 in the nervous system and relative contributions of gain- or loss- of function to the pathogenesis of CIZ1-associated dystonia remain indefinite. Using relative quantitative reverse transcriptase-PCR, cerebellum showed the highest expression levels of Ciz1 in adult mouse brain, over two fold higher than liver, and higher than striatum, midbrain and cerebral cortex. Overall, neural expression of Ciz1 increased with postnatal age. A Ciz1 gene-trap knock-out (KO) mouse model (Ciz1(-/-)) was generated to examine the functional role(s) of CIZ1 in the sensorimotor nervous system and contributions of CIZ1 to cell-cycle control in the mammalian brain. Ciz1 transcripts were absent in Ciz1(-/-) mice and reduced by approximately 50% in Ciz1(+/-) mice. Ciz1(-/-) mice were fertile but smaller than wild-type (WT) littermates. Ciz1(-/-) mice did not manifest dystonia, but exhibited mild motoric abnormalities on balance, open-field activity, and gait. To determine the effects of germline KO of Ciz1 on whole-genome gene expression in adult brain, total RNA from mouse cerebellum was harvested from 6 10-month old Ciz1(-/-) mice and 6 age- and gender- matched WT littermates for whole-genome gene expression analysis. Based on whole-genome gene-expression analyses, genes involved in cellular movement, cell development, cellular growth, cellular morphology and cell-to-cell signaling and interaction were up-regulated in Ciz1(-/-) mice. The top up-regulated pathways were metabolic and cytokine-cytokine receptor interactions. Down-regulated genes were involved in cell cycle, cellular development, cell death and survival, gene expression and cell morphology. Down-regulated networks included those related to metabolism, focal adhesion, neuroactive ligand-receptor interaction, and MAPK signaling. Based on pathway analyses, transcription factor 7-like 2 (TCF7L2), a member of the Wnt/β-catenin signaling pathway, was a major hub for down-regulated genes, whereas NF-κB was a major hub for up-regulated genes. In aggregate, these data suggest that CIZ1 may be involved in the post-mitotic differentiation of neurons in response to external signals and changes in gene expression may compensate, in part, for CIZ1 deficiency in our Ciz1(-/-) mouse model. Although CIZ1 deficiency was associated with mild motor abnormalities, germline loss of Ciz1 was not associated with dystonia on the C57BL/6J background.
Collapse
Affiliation(s)
- Jianfeng Xiao
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Satya R Vemula
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yi Xue
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Mohammad M Khan
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Korah P Kuruvilla
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Esther M Marquez-Lona
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Madison R Cobb
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Mark S LeDoux
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
18
|
Domingo A, Erro R, Lohmann K. Novel Dystonia Genes: Clues on Disease Mechanisms and the Complexities of High-Throughput Sequencing. Mov Disord 2016; 31:471-7. [PMID: 26991507 DOI: 10.1002/mds.26600] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 02/08/2016] [Accepted: 02/11/2016] [Indexed: 12/24/2022] Open
Abstract
Dystonia is a genetically heterogenous disease and a prototype disorder where next-generation sequencing has facilitated the identification of new pathogenic genes. This includes the first two genes linked to recessively inherited isolated dystonia, that is, HPCA (hippocalcin) and COL6A3 (collagen VI alpha 3). These genes are proposed to underlie cases of the so-called DYT2-like dystonia, while also reiterating two distinct pathways in dystonia pathogenesis. First, deficiency in HPCA function is thought to alter calcium homeostasis, a mechanism that has previously been forwarded for CACNA1A and ANO3. The novel myoclonus-dystonia genes KCTD17 and CACNA1B also implicate abnormal calcium signaling in dystonia. Second, the phenotype in COL6A3-loss-of-function zebrafish models argues for a neurodevelopmental defect, which has previously been suggested as a possible biological mechanism for THAP1, TOR1A, and TAF1 based on expression data. The newly reported myoclonus-dystonia gene, RELN, plays also a role in the formation of brain structures. Defects in neurodevelopment likewise seem to be a recurrent scheme underpinning mainly complex dystonias, for example those attributable to biallelic mutations in GCH1, TH, SPR, or to heterozygous TUBB4A mutations. To date, it remains unclear whether dystonia is a common phenotypic outcome of diverse underlying disease mechanisms, or whether the different genetic causes converge in a single pathway. Importantly, the relevance of pathways highlighted by novel dystonia genes identified by high-throughput sequencing depends on the confirmation of mutation pathogenicity in subsequent genetic and functional studies. However, independent, careful validation of genetic findings lags behind publications of newly identified genes. We conclude with a discussion on the characteristics of true-positive reports.
Collapse
Affiliation(s)
- Aloysius Domingo
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Roberto Erro
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, United Kingdom
- Dipartimento di Scienze Neurologiche e del Movimento, Università di Verona, Verona, Italy
| | - Katja Lohmann
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| |
Collapse
|
19
|
Ruiz M, Perez-Garcia G, Ortiz-Virumbrales M, Méneret A, Morant A, Kottwitz J, Fuchs T, Bonet J, Gonzalez-Alegre P, Hof PR, Ozelius LJ, Ehrlich ME. Abnormalities of motor function, transcription and cerebellar structure in mouse models of THAP1 dystonia. Hum Mol Genet 2015; 24:7159-70. [PMID: 26376866 DOI: 10.1093/hmg/ddv384] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/14/2015] [Indexed: 01/07/2023] Open
Abstract
DYT6 dystonia is caused by mutations in THAP1 [Thanatos-associated (THAP) domain-containing apoptosis-associated protein] and is autosomal dominant and partially penetrant. Like other genetic primary dystonias, DYT6 patients have no characteristic neuropathology, and mechanisms by which mutations in THAP1 cause dystonia are unknown. Thap1 is a zinc-finger transcription factor, and most pathogenic THAP1 mutations are missense and are located in the DNA-binding domain. There are also nonsense mutations, which act as the equivalent of a null allele because they result in the generation of small mRNA species that are likely rapidly degraded via nonsense-mediated decay. The function of Thap1 in neurons is unknown, but there is a unique, neuronal 50-kDa Thap1 species, and Thap1 levels are auto-regulated on the mRNA level. Herein, we present the first characterization of two mouse models of DYT6, including a pathogenic knockin mutation, C54Y and a null mutation. Alterations in motor behaviors, transcription and brain structure are demonstrated. The projection neurons of the deep cerebellar nuclei are especially altered. Abnormalities vary according to genotype, sex, age and/or brain region, but importantly, overlap with those of other dystonia mouse models. These data highlight the similarities and differences in age- and cell-specific effects of a Thap1 mutation, indicating that the pathophysiology of THAP1 mutations should be assayed at multiple ages and neuronal types and support the notion of final common pathways in the pathophysiology of dystonia arising from disparate mutations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Pedro Gonzalez-Alegre
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Patrick R Hof
- Department of Neurosciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA and
| | - Laurie J Ozelius
- Department of Genetics and Genomic Sciences, Department of Neurology
| | - Michelle E Ehrlich
- Department of Pediatrics, Department of Genetics and Genomic Sciences, Department of Neurology,
| |
Collapse
|
20
|
Antelmi E, Erro R, Pisani A, Mencacci N, Bhatia KP. Persistent chorea in DYT6, due to anticholinergic therapy. Parkinsonism Relat Disord 2015; 21:1282-3. [PMID: 26275586 DOI: 10.1016/j.parkreldis.2015.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 07/28/2015] [Accepted: 07/31/2015] [Indexed: 01/11/2023]
Affiliation(s)
- E Antelmi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy; Sobell Department of Motor Neuroscience and Movement Disorders, University College London (UCL) Institute of Neurology, London, UK.
| | - R Erro
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London (UCL) Institute of Neurology, London, UK
| | - A Pisani
- Department of Systems Medicine, University of Rome "Tor Vergata", Italy; Laboratory of Neurophysiology and Synaptic Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - N Mencacci
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London (UCL) Institute of Neurology, London, UK
| | - K P Bhatia
- Sobell Department of Motor Neuroscience and Movement Disorders, University College London (UCL) Institute of Neurology, London, UK
| |
Collapse
|
21
|
Abstract
The dystonias are a group of disorders characterized by excessive involuntary muscle contractions leading to abnormal postures and/or repetitive movements. A careful assessment of the clinical manifestations is helpful for identifying syndromic patterns that focus diagnostic testing on potential causes. If a cause is identified, specific etiology-based treatments may be available. In most cases, a specific cause cannot be identified, and treatments are based on symptoms. Treatment options include counseling, education, oral medications, botulinum toxin injections, and several surgical procedures. A substantial reduction in symptoms and improved quality of life is achieved in most patients by combining these options.
Collapse
Affiliation(s)
- H A Jinnah
- Department of Neurology, Emory University School of Medicine, 6300 Woodruff Memorial Research Building, 101 Woodruff Circle, Emory University, Atlanta, GA 30322, USA; Department of Human Genetics, Emory University School of Medicine, 6300 Woodruff Memorial Research Building, 101 Woodruff Circle, Emory University, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, 6300 Woodruff Memorial Research Building, 101 Woodruff Circle, Emory University, Atlanta, GA 30322, USA.
| | - Stewart A Factor
- Department of Neurology, Emory University School of Medicine, 6300 Woodruff Memorial Research Building, 101 Woodruff Circle, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
22
|
Recessive mutations in the α3 (VI) collagen gene COL6A3 cause early-onset isolated dystonia. Am J Hum Genet 2015; 96:883-93. [PMID: 26004199 DOI: 10.1016/j.ajhg.2015.04.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 04/16/2015] [Indexed: 12/13/2022] Open
Abstract
Isolated dystonia is a disorder characterized by involuntary twisting postures arising from sustained muscle contractions. Although autosomal-dominant mutations in TOR1A, THAP1, and GNAL have been found in some cases, the molecular mechanisms underlying isolated dystonia are largely unknown. In addition, although emphasis has been placed on dominant isolated dystonia, the disorder is also transmitted as a recessive trait, for which no mutations have been defined. Using whole-exome sequencing in a recessive isolated dystonia-affected kindred, we identified disease-segregating compound heterozygous mutations in COL6A3, a collagen VI gene associated previously with muscular dystrophy. Genetic screening of a further 367 isolated dystonia subjects revealed two additional recessive pedigrees harboring compound heterozygous mutations in COL6A3. Strikingly, all affected individuals had at least one pathogenic allele in exon 41, including an exon-skipping mutation that induced an in-frame deletion. We tested the hypothesis that disruption of this exon is pathognomonic for isolated dystonia by inducing a series of in-frame deletions in zebrafish embryos. Consistent with our human genetics data, suppression of the exon 41 ortholog caused deficits in axonal outgrowth, whereas suppression of other exons phenocopied collagen deposition mutants. All recessive mutation carriers demonstrated early-onset segmental isolated dystonia without muscular disease. Finally, we show that Col6a3 is expressed in neurons, with relevant mRNA levels detectable throughout the adult mouse brain. Taken together, our data indicate that loss-of-function mutations affecting a specific region of COL6A3 cause recessive isolated dystonia with underlying neurodevelopmental deficits and highlight the brain extracellular matrix as a contributor to dystonia pathogenesis.
Collapse
|
23
|
Mencacci N, Rubio-Agusti I, Zdebik A, Asmus F, Ludtmann M, Ryten M, Plagnol V, Hauser AK, Bandres-Ciga S, Bettencourt C, Forabosco P, Hughes D, Soutar M, Peall K, Morris H, Trabzuni D, Tekman M, Stanescu H, Kleta R, Carecchio M, Zorzi G, Nardocci N, Garavaglia B, Lohmann E, Weissbach A, Klein C, Hardy J, Pittman A, Foltynie T, Abramov A, Gasser T, Bhatia K, Wood N. A missense mutation in KCTD17 causes autosomal dominant myoclonus-dystonia. Am J Hum Genet 2015; 96:938-47. [PMID: 25983243 DOI: 10.1016/j.ajhg.2015.04.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/13/2015] [Indexed: 12/25/2022] Open
Abstract
Myoclonus-dystonia (M-D) is a rare movement disorder characterized by a combination of non-epileptic myoclonic jerks and dystonia. SGCE mutations represent a major cause for familial M-D being responsible for 30%-50% of cases. After excluding SGCE mutations, we identified through a combination of linkage analysis and whole-exome sequencing KCTD17 c.434 G>A p.(Arg145His) as the only segregating variant in a dominant British pedigree with seven subjects affected by M-D. A subsequent screening in a cohort of M-D cases without mutations in SGCE revealed the same KCTD17 variant in a German family. The clinical presentation of the KCTD17-mutated cases was distinct from the phenotype usually observed in M-D due to SGCE mutations. All cases initially presented with mild myoclonus affecting the upper limbs. Dystonia showed a progressive course, with increasing severity of symptoms and spreading from the cranio-cervical region to other sites. KCTD17 is abundantly expressed in all brain regions with the highest expression in the putamen. Weighted gene co-expression network analysis, based on mRNA expression profile of brain samples from neuropathologically healthy individuals, showed that KCTD17 is part of a putamen gene network, which is significantly enriched for dystonia genes. Functional annotation of the network showed an over-representation of genes involved in post-synaptic dopaminergic transmission. Functional studies in mutation bearing fibroblasts demonstrated abnormalities in endoplasmic reticulum-dependent calcium signaling. In conclusion, we demonstrate that the KCTD17 c.434 G>A p.(Arg145His) mutation causes autosomal dominant M-D. Further functional studies are warranted to further characterize the nature of KCTD17 contribution to the molecular pathogenesis of M-D.
Collapse
|
24
|
Abstract
OPINION STATEMENT Dystonia is a movement disorder caused by diverse etiologies. Its treatment in children is particularly challenging due to the complexity of the development of the nervous system from birth to young adulthood. The treatment options of childhood dystonia include several oral pharmaceutical agents, botulinum toxin injections, and deep brain stimulation (DBS) therapy. The choice of drug therapy relies on the suspected etiology of the dystonia and the adverse effect profile of the drugs. Dystonic syndromes with known etiologies may require specific interventions, but most dystonias are treated by trying serially a handful of medications starting with those with the best risk/benefit profile. In conjunction to drug therapy, botulinum toxin injections may be used to target a problematic group dystonic muscles. The maximal botulinum toxin dose is limited by the weight of the child, therefore limiting the number of the muscles amenable to such treatment. When drugs and botulinum toxin injections fail to control the child's disabling dystonia, DBS therapy may be offered as a last remedy. Delivering optimal DBS therapy to children with dystonia requires a multidisciplinary team of experienced pediatric neurosurgeons, neurologists, and nurses to select adequate candidates, perform this delicate stereotactic procedure, and optimize DBS delivery. Even in the best hands, the response of childhood dystonia to DBS therapy varies greatly. Future therapy of childhood dystonia will parallel the advancement of knowledge of the pathophysiology of dystonic syndromes and the development of clinical and research tools for their study.
Collapse
Affiliation(s)
- Samer D Tabbal
- Department of Neurology, American University of Beirut, Riad El-Solh, PO Box 11-0236, Beirut, 1107 2020, Lebanon,
| |
Collapse
|
25
|
Abstract
Dystonia, a common and genetically heterogeneous neurological disorder, was recently defined as "a movement disorder characterized by sustained or intermittent muscle contractions causing abnormal, often repetitive, movements, postures, or both." Via the application of whole-exome sequencing, the genetic landscape of dystonia and closely related movement disorders is becoming exposed. In particular, several "novel" genetic causes have been causally associated with dystonia or dystonia-related disorders over the past 2 years. These genes include PRRT2 (DYT10), CIZ1 (DYT23), ANO3 (DYT24), GNAL (DYT25), and TUBB4A (DYT4). Despite these advances, major gaps remain in identifying the genetic origins for most cases of adult-onset isolated dystonia. Furthermore, model systems are needed to study the biology of PRRT2, CIZ1, ANO3, Gαolf, and TUBB4A in the context of dystonia. This review focuses on these recent additions to the family of dystonia genes, genotype-phenotype correlations, and possible cellular contributions of the encoded proteins to the development of dystonia.
Collapse
Affiliation(s)
- Jianfeng Xiao
- Department of Neurology, University of Tennessee Health Science Center, 855 Monroe Avenue, Link Building Suite 415, Memphis, TN, 38163, USA,
| | | | | |
Collapse
|
26
|
Striatal cholinergic dysfunction as a unifying theme in the pathophysiology of dystonia. Prog Neurobiol 2015; 127-128:91-107. [PMID: 25697043 DOI: 10.1016/j.pneurobio.2015.02.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 02/05/2015] [Accepted: 02/07/2015] [Indexed: 01/06/2023]
Abstract
Dystonia is a movement disorder of both genetic and non-genetic causes, which typically results in twisted posturing due to abnormal muscle contraction. Evidence from dystonia patients and animal models of dystonia indicate a crucial role for the striatal cholinergic system in the pathophysiology of dystonia. In this review, we focus on striatal circuitry and the centrality of the acetylcholine system in the function of the basal ganglia in the control of voluntary movement and ultimately clinical manifestation of movement disorders. We consider the impact of cholinergic interneurons (ChIs) on dopamine-acetylcholine interactions and examine new evidence for impairment of ChIs in dysfunction of the motor systems producing dystonic movements, particularly in animal models. We have observed paradoxical excitation of ChIs in the presence of dopamine D2 receptor agonists and impairment of striatal synaptic plasticity in a mouse model of DYT1 dystonia, which are improved by administration of recently developed M1 receptor antagonists. These findings have been confirmed across multiple animal models of DYT1 dystonia and may represent a common endophenotype by which to investigate dystonia induced by other types of genetic and non-genetic causes and to investigate the potential effectiveness of pharmacotherapeutics and other strategies to improve dystonia.
Collapse
|
27
|
Abstract
There has been considerable progress in our understanding of dystonia over the last century. Growing recognition of dystonia has enhanced awareness of its diverse motor phenomenology and brought attention to the importance that nonmotor features may play in this disorder, once considered to be purely motor. Using the latest technologies in human genetics, new genetic links are being discovered at an ever-quickening pace and expanding our knowledge of the disorder's complex pathogenesis. Furthermore, as we gain clearer insight into the pathophysiology of dystonia and an appreciation of the involvement of dysfunction outside the basal ganglia, dystonia has been increasingly viewed as a network disorder. Here we briefly discuss some of the recent noteworthy advances.
Collapse
Affiliation(s)
- Brian D Berman
- Department of Neurology (BDB), University of Colorado Denver, Aurora, CO; and Departments of Neurology, Human Genetics, and Pediatrics (HAJ), Emory University, Atlanta, GA
| | - H A Jinnah
- Department of Neurology (BDB), University of Colorado Denver, Aurora, CO; and Departments of Neurology, Human Genetics, and Pediatrics (HAJ), Emory University, Atlanta, GA
| |
Collapse
|
28
|
Groen JL, Ritz K, Jalalzadeh H, van der Salm SM, Jongejan A, Mook OR, Haagmans MA, Zwinderman AH, Motazacker MM, Hennekam RC, Baas F, Tijssen MA. RELN
rare variants in myoclonus‐dystonia. Mov Disord 2015; 30:415-9. [DOI: 10.1002/mds.26070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/30/2014] [Accepted: 10/05/2014] [Indexed: 12/25/2022] Open
Affiliation(s)
- Justus L. Groen
- Department of NeurologyAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
- Department of Genome AnalysisAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Katja Ritz
- Department of Genome AnalysisAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Hamid Jalalzadeh
- Department of NeurologyAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Sandra M.A. van der Salm
- Department of NeurologyAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Aldo Jongejan
- Department of Clinical Epidemiology and Bio‐StatisticsAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Olaf R. Mook
- Department of Clinical GeneticsAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Martin A. Haagmans
- Department of Clinical GeneticsAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Aeilko H. Zwinderman
- Department of Clinical Epidemiology and Bio‐StatisticsAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Mahdi M. Motazacker
- Department of Clinical GeneticsAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Raoul C. Hennekam
- Department of PediatricsAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | - Frank Baas
- Department of Genome AnalysisAcademic Medical Center, University of AmsterdamAmsterdam The Netherlands
| | | |
Collapse
|
29
|
Jinnah H, Albanese A. The New Classification System for the Dystonias: Why Was it Needed and How was it Developed? Mov Disord Clin Pract 2014; 1:280-284. [PMID: 25485288 PMCID: PMC4254809 DOI: 10.1002/mdc3.12100] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 09/08/2014] [Accepted: 09/09/2014] [Indexed: 12/18/2022] Open
Affiliation(s)
- H.A. Jinnah
- Departments of Neurology, Human Genetics and PediatricsEmory UniversityAtlantaGeorgia30322USA
| | - Alberto Albanese
- Department of NeurologyCatholic UniversityMilanItaly
- Department of NeurologyCarlo Besta Institute of NeurologyMilanItaly
| |
Collapse
|
30
|
Ortiz-Virumbrales M, Ruiz M, Hone E, Dolios G, Wang R, Morant A, Kottwitz J, Ozelius LJ, Gandy S, Ehrlich ME. Dystonia type 6 gene product Thap1: identification of a 50 kDa DNA-binding species in neuronal nuclear fractions. Acta Neuropathol Commun 2014; 2:139. [PMID: 25231164 PMCID: PMC4177242 DOI: 10.1186/s40478-014-0139-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 09/05/2014] [Indexed: 01/04/2023] Open
Abstract
Mutations in THAP1 result in dystonia type 6, with partial penetrance and variable phenotype. The goal of this study was to examine the nature and expression pattern of the protein product(s) of the Thap1 transcription factor (DYT6 gene) in mouse neurons, and to study the regional and developmental distribution, and subcellular localization of Thap1 protein. The goal was accomplished via overexpression and knock-down of Thap1 in the HEK293T cell line and in mouse striatal primary cultures and western blotting of embryonic Thap1-null tissue. The endogenous and transduced Thap1 isoforms were characterized using three different commercially available anti-Thap1 antibodies and validated by immunoprecipitation and DNA oligonucleotide affinity chromatography. We identified multiple, novel Thap1 species of apparent Mr 32 kDa, 47 kDa, and 50–52 kDa in vitro and in vivo, and verified the previously identified species at 29–30 kDa in neurons. The Thap1 species at the 50 kDa size range was exclusively detected in murine brain and testes and were located in the nuclear compartment. Thus, in addition to the predicted 25 kDa apparent Mr, we identified Thap1 species with greater apparent Mr that we speculate may be a result of posttranslational modifications. The neural localization of the 50 kDa species and its nuclear compartmentalization suggests that these may be key Thap1 species controlling neuronal gene transcription. Dysfunction of the neuronal 50 kDa species may therefore be implicated in the pathogenesis of DYT6.
Collapse
|
31
|
Genetic animal models of dystonia: common features and diversities. Prog Neurobiol 2014; 121:91-113. [PMID: 25034123 DOI: 10.1016/j.pneurobio.2014.07.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 06/06/2014] [Accepted: 07/03/2014] [Indexed: 01/13/2023]
Abstract
Animal models are pivotal for studies of pathogenesis and treatment of disorders of the central nervous system which in its complexity cannot yet be modeled in vitro or using computer simulations. The choice of a specific model to test novel therapeutic strategies for a human disease should be based on validity of the model for the approach: does the model reflect symptoms, pathogenesis and treatment response present in human patients? In the movement disorder dystonia, prior to the availability of genetically engineered mice, spontaneous mutants were chosen based on expression of dystonic features, including abnormal muscle contraction, movements and postures. Recent discovery of a number of genes and gene products involved in dystonia initiated research on pathogenesis of the disorder, and the creation of novel models based on gene mutations. Here we present a review of current models of dystonia, with a focus on genetic rodent models, which will likely be first choice in the future either for pathophysiological or for preclinical drug testing or both. In order to help selection of a model depending on expression of a specific feature of dystonia, this review is organized by symptoms and current knowledge of pathogenesis of dystonia. We conclude that albeit there is increasing need for research on pathogenesis of the disease and development of improved models, current models do replicate features of dystonia and are useful tools to develop urgently demanded treatment for this debilitating disorder.
Collapse
|
32
|
Vemula SR, Xiao J, Zhao Y, Bastian RW, Perlmutter JS, Racette BA, Paniello RC, Wszolek ZK, Uitti RJ, Van Gerpen JA, Hedera P, Truong DD, Blitzer A, Rudzińska M, Momčilović D, Jinnah HA, Frei K, Pfeiffer RF, LeDoux MS. A rare sequence variant in intron 1 of THAP1 is associated with primary dystonia. Mol Genet Genomic Med 2014; 2:261-72. [PMID: 24936516 PMCID: PMC4049367 DOI: 10.1002/mgg3.67] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/31/2013] [Accepted: 01/03/2014] [Indexed: 12/16/2022] Open
Abstract
Although coding variants in THAP1 have been causally associated with primary dystonia, the contribution of noncoding variants remains uncertain. Herein, we examine a previously identified Intron 1 variant (c.71+9C>A, rs200209986). Among 1672 subjects with mainly adult-onset primary dystonia, 12 harbored the variant in contrast to 1/1574 controls (P < 0.01). Dystonia classification included cervical dystonia (N = 3), laryngeal dystonia (adductor subtype, N = 3), jaw-opening oromandibular dystonia (N = 1), blepharospasm (N = 2), and unclassified (N = 3). Age of dystonia onset ranged from 25 to 69 years (mean = 54 years). In comparison to controls with no identified THAP1 sequence variants, the c.71+9C>A variant was associated with an elevated ratio of Isoform 1 (NM_018105) to Isoform 2 (NM_199003) in leukocytes. In silico and minigene analyses indicated that c.71+9C>A alters THAP1 splicing. Lymphoblastoid cells harboring the c.71+9C>A variant showed extensive apoptosis with relatively fewer cells in the G2 phase of the cell cycle. Differentially expressed genes from lymphoblastoid cells revealed that the c.71+9C>A variant exerts effects on DNA synthesis, cell growth and proliferation, cell survival, and cytotoxicity. In aggregate, these data indicate that THAP1 c.71+9C>A is a risk factor for adult-onset primary dystonia.
Collapse
Affiliation(s)
- Satya R Vemula
- Departments of Neurology and Anatomy & Neurobiology, University of Tennessee Health Science Center Memphis, Tennessee, 38163
| | - Jianfeng Xiao
- Departments of Neurology and Anatomy & Neurobiology, University of Tennessee Health Science Center Memphis, Tennessee, 38163
| | - Yu Zhao
- Departments of Neurology and Anatomy & Neurobiology, University of Tennessee Health Science Center Memphis, Tennessee, 38163
| | | | - Joel S Perlmutter
- Department of Neurology, Washington University School of Medicine St. Louis, Missouri
| | - Brad A Racette
- Department of Neurology, Washington University School of Medicine St. Louis, Missouri
| | - Randal C Paniello
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine St. Louis, Missouri
| | | | - Ryan J Uitti
- Department of Neurology, Mayo Clinic Jacksonville, Florida, 32224
| | - Jay A Van Gerpen
- Department of Neurology, Mayo Clinic Jacksonville, Florida, 32224
| | - Peter Hedera
- Department of Neurology, Vanderbilt University Nashville, Tennessee
| | - Daniel D Truong
- Parkinson's & Movement Disorder Institute Fountain Valley, California, 92708
| | - Andrew Blitzer
- New York Center for Voice and Swallowing Disorders New York, New York
| | - Monika Rudzińska
- Department of Neurology, Jagiellonian University Medical College in Krakow Kraków, Poland
| | - Dragana Momčilović
- Clinic for Child Neurology and Psychiatry, Medical Faculty University of Belgrade Belgrade, Serbia
| | - Hyder A Jinnah
- Departments of Neurology, Human Genetics, and Pediatrics, School of Medicine, Emory University Atlanta, Georgia, 30322
| | - Karen Frei
- Department of Neurology, Loma Linda University Health System Loma Linda, California, 92354
| | - Ronald F Pfeiffer
- Departments of Neurology and Anatomy & Neurobiology, University of Tennessee Health Science Center Memphis, Tennessee, 38163
| | - Mark S LeDoux
- Departments of Neurology and Anatomy & Neurobiology, University of Tennessee Health Science Center Memphis, Tennessee, 38163
| |
Collapse
|
33
|
Dystonia as a network disorder: what is the role of the cerebellum? Neuroscience 2013; 260:23-35. [PMID: 24333801 DOI: 10.1016/j.neuroscience.2013.11.062] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 11/20/2013] [Accepted: 11/20/2013] [Indexed: 01/02/2023]
Abstract
The dystonias are a group of disorders defined by sustained or intermittent muscle contractions that result in involuntary posturing or repetitive movements. There are many different clinical manifestations and causes. Although they traditionally have been ascribed to dysfunction of the basal ganglia, recent evidence has suggested dysfunction may originate from other regions, particularly the cerebellum. This recent evidence has led to an emerging view that dystonia is a network disorder that involves multiple brain regions. The new network model for the pathogenesis of dystonia has raised many questions, particularly regarding the role of the cerebellum. For example, if dystonia may arise from cerebellar dysfunction, then why are there no cerebellar signs in dystonia? Why are focal cerebellar lesions or degenerative cerebellar disorders more commonly associated with ataxia rather than dystonia? Why is dystonia more commonly associated with basal ganglia lesions rather than cerebellar lesions? Can answers obtained from animals be extrapolated to humans? Is there any evidence that the cerebellum is not involved? Finally, what is the practical value of this new model of pathogenesis for the neuroscientist and clinician? This article explores potential answers to these questions.
Collapse
|
34
|
Jinnah HA, DeLong M, Hallett M. The dystonias: past, present, and future. Mov Disord 2013; 28:849-50. [PMID: 23893441 PMCID: PMC3787865 DOI: 10.1002/mds.25564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 11/06/2022] Open
Affiliation(s)
- H. A. Jinnah
- Departments of Neurology, Human Genetics & Pediatrics, Emory University, Atlanta GA, USA
| | - Mahlon DeLong
- Department of Neurology, Pediatrics & Psychiatry, Emory University, Atlanta GA, USA
| | - Mark Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda MD, USA
| |
Collapse
|
35
|
Jinnah HA, Berardelli A, Comella C, Defazio G, Delong MR, Factor S, Galpern WR, Hallett M, Ludlow CL, Perlmutter JS, Rosen AR. The focal dystonias: current views and challenges for future research. Mov Disord 2013; 28:926-43. [PMID: 23893450 PMCID: PMC3733486 DOI: 10.1002/mds.25567] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 11/11/2022] Open
Abstract
The most common forms of dystonia are those that develop in adults and affect a relatively isolated region of the body. Although these adult-onset focal dystonias are most prevalent, knowledge of their etiologies and pathogenesis has lagged behind some of the rarer generalized dystonias, in which the identification of genetic defects has facilitated both basic and clinical research. This summary provides a brief review of the clinical manifestations of the adult-onset focal dystonias, focusing attention on less well understood clinical manifestations that need further study. It also provides a simple conceptual model for the similarities and differences among the different adult-onset focal dystonias as a rationale for lumping them together as a class of disorders while at the same time splitting them into subtypes. The concluding section outlines some of the most important research questions for the future. Answers to these questions are critical for advancing our understanding of this group of disorders and for developing novel therapeutics.
Collapse
Affiliation(s)
- H A Jinnah
- Department of Neurology, Emory University, Atlanta, Georgia 30322, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|