1
|
Olivera C, Boscolo O, Dobrecky C, Ortega CA, Favier LS, Cianchino VA, Flor S, Lucangioli S. Development and Characterization of Trihexyphenidyl Orodispersible Minitablets: A Challenge to Fill the Therapeutic Gap in Neuropediatrics. Pharmaceutics 2024; 17:5. [PMID: 39861657 PMCID: PMC11769368 DOI: 10.3390/pharmaceutics17010005] [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: 11/04/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Trihexyphenidyl (THP) has been widely used for over three decades as pediatric pharmacotherapy in patients affected by segmental and generalized dystonia. In order to achieve effective and safe pharmacotherapy for this population, new formulations are needed. Objective: The aim of this work is the development of trihexyphenidyl orodispersible minitablets (ODMTs) for pediatric use. Methods: Six different excipients were tested as diluents. The properties of powder mixtures were evaluated before direct compression and pharmacotechnical tests were performed on the final formulation. The determination of the API content, uniformity of dosage, and physicochemical stability studies were analyzed by an HPLC-UV method. Results: The developed ODMTs met pharmacopeia specifications for content, hardness, friability, disintegration, and dissolution tests. The physicochemical stability study performed over 18 months shows that API content remains within 90.0-110.0% at least for this period. Conclusions: These ODMTs will allow efficient, safe, and high-quality pharmacotherapy.
Collapse
Affiliation(s)
- Camila Olivera
- Departamento de Tecnología Farmacéutica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina; (C.O.); (S.F.)
- Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Buenos Aires C1414AAD, Argentina
| | - Oriana Boscolo
- Departamento de Tecnología Farmacéutica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina; (C.O.); (S.F.)
- Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina
| | - Cecilia Dobrecky
- Departamento de Tecnología Farmacéutica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina; (C.O.); (S.F.)
- Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina
- Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina
| | - Claudia A. Ortega
- Área de Farmacotecnia, Ética y Legislación Farmacéutica, Departamento de Farmacia, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis D5700HHW, Argentina
| | - Laura S. Favier
- Área de Farmacotecnia, Ética y Legislación Farmacéutica, Departamento de Farmacia, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis D5700HHW, Argentina
| | - Valeria A. Cianchino
- Área de Farmacotecnia, Ética y Legislación Farmacéutica, Departamento de Farmacia, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis D5700HHW, Argentina
| | - Sabrina Flor
- Departamento de Tecnología Farmacéutica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina; (C.O.); (S.F.)
- Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Buenos Aires C1414AAD, Argentina
| | - Silvia Lucangioli
- Departamento de Tecnología Farmacéutica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina; (C.O.); (S.F.)
- Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Buenos Aires C1414AAD, Argentina
| |
Collapse
|
2
|
LeWitt PA, Hong L, Moehle MS. Anticholinergic drugs for parkinsonism and other movement disorders. J Neural Transm (Vienna) 2024; 131:1481-1494. [PMID: 38904792 DOI: 10.1007/s00702-024-02799-7] [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: 02/07/2024] [Accepted: 06/02/2024] [Indexed: 06/22/2024]
Abstract
Anticholinergic (AC) drugs, a medication class that acts by blocking nicotinic and muscarinic acetylcholine receptors, were first utilized for therapeutic purposes in the mid-19th century. Initial applications were as symptomatic therapy for Parkinson disease (PD), a practice continuing to the present. Initially, the AC drugs used were naturally-occurring plant compounds. Synthetic AC drugs were developed in the late 1940s and predominated in neurological therapeutics. Until the advent of pharmaceuticals acting upon striatal dopaminergic motor pathways, AC drugs provided the only effective means for lessening tremors and other clinical problems of the PD patient. However, because dopaminergic compounds are so effective at meeting the needs of the typical PD patient, AC medications are far less utilized by clinicians today. In recent years, there has been only a few investigations of AC drugs as neurological treatments. This review will revisit the clinical landscape of AC pharmacology and application for movement disorders along with recent research in search of improving therapeutics with AC drugs.
Collapse
Affiliation(s)
- Peter A LeWitt
- Departments of Neurology, Wayne State University School of Medicine and Henry Ford Hospital, 8-D 4201 St. Antoine Street, Detroit, MI, 48201, USA.
| | - Luke Hong
- The Department of Neurology, University of South Florida, Tampa, FL, USA
| | - Mark S Moehle
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, USA
| |
Collapse
|
3
|
Katragadda P, Holla VV, Kamble N, Yadav R, Pal PK. Haloperidol in Managing DYT-TOR1A Dystonia: Unveiling a Dramatic Therapeutic Response. J Mov Disord 2024; 17:342-344. [PMID: 38589017 PMCID: PMC11300396 DOI: 10.14802/jmd.24029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/19/2024] [Accepted: 04/05/2024] [Indexed: 04/10/2024] Open
Affiliation(s)
- Pavankumar Katragadda
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Vikram V. Holla
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Nitish Kamble
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| |
Collapse
|
4
|
Abstract
Dystonia is a clinically and genetically highly heterogeneous neurological disorder characterized by abnormal movements and postures caused by involuntary sustained or intermittent muscle contractions. A number of groundbreaking genetic and molecular insights have recently been gained. While they enable genetic testing and counseling, their translation into new therapies is still limited. However, we are beginning to understand shared pathophysiological pathways and molecular mechanisms. It has become clear that dystonia results from a dysfunctional network involving the basal ganglia, cerebellum, thalamus, and cortex. On the molecular level, more than a handful of, often intertwined, pathways have been linked to pathogenic variants in dystonia genes, including gene transcription during neurodevelopment (e.g., KMT2B, THAP1), calcium homeostasis (e.g., ANO3, HPCA), striatal dopamine signaling (e.g., GNAL), endoplasmic reticulum stress response (e.g., EIF2AK2, PRKRA, TOR1A), autophagy (e.g., VPS16), and others. Thus, different forms of dystonia can be molecularly grouped, which may facilitate treatment development in the future.
Collapse
Affiliation(s)
- Mirja Thomsen
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Lara M Lange
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| |
Collapse
|
5
|
Kumar S, Shankar Kaushik J, Verma S, Dabla S. Gabapentin as Add-on Therapy to Trihexyphenidyl in Children with Dyskinetic Cerebral Palsy: A Randomized, Controlled Trial. Indian J Pediatr 2023; 90:873-879. [PMID: 35867274 DOI: 10.1007/s12098-022-04265-2] [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: 01/31/2022] [Accepted: 04/29/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To compare the efficacy of gabapentin as add-on therapy to trihexyphenidyl in the treatment of children with dyskinetic cerebral palsy (CP). METHODS An open-labelled, randomized, controlled trial was conducted among children aged 3-9 y with dyskinetic CP [Gross Motor Functional Classification System (GMFCS) 4-5]. Participants were assigned into two groups: gabapentin with trihexyphenidyl (n = 30) and trihexyphenidyl alone (n = 30). Dyskinesia Impairment Scale (DIS), Dystonia Severity Assessment Plan (DSAP), and International Classification of Functioning, Disability, and Health-Children and Youth Version (ICF-CY) were measured at baseline, 4 and 12 wk. RESULTS There was significant reduction in baseline dystonia in both the groups (DIS: p < 0.001; DSAP: p = 0.007; ICF-CY: p < 0.001) but when data were compared between the groups, there was no significant difference in the severity of dystonia at 4 wk and at 12 wk (DIS: p = 0.09; DSAP: p = 0.49; ICF-CY: p = 0.25). Constipation was the commonest side effect observed in both the groups [3 (11.5%) vs. 4 (14.3%)]. CONCLUSION Trihexyphenidyl alone is as effective as combination of gabapentin with trihexyphenidyl in decreasing the severity of dystonia at 12 wk. Hence, there is no added benefit of gabapentin as add-on therapy for dystonia among children with dyskinetic CP. TRIAL REGISTRATION CTRI/2019/04/018603.
Collapse
Affiliation(s)
- Sonu Kumar
- Department of Pediatrics, SGT University, Gurugram, Haryana, India
| | - Jaya Shankar Kaushik
- Department of Pediatrics, Pt. B. D. Sharma Post Graduate Institute of Medical Sciences, Rohtak, Haryana, 124001, India.
| | - Savita Verma
- Department of Pharmacology, Pt. B. D. Sharma Post Graduate Institute of Medical Sciences, Rohtak, Haryana, India
| | - Surekha Dabla
- Department of Neurology, Pt. B. D. Sharma Post Graduate Institute of Medical Sciences, Rohtak, Haryana, India
| |
Collapse
|
6
|
Wilkes BJ, Adury RZ, Berryman D, Concepcion LR, Liu Y, Yokoi F, Maugee C, Li Y, Vaillancourt DE. Cell-specific Dyt1 ∆GAG knock-in to basal ganglia and cerebellum reveal differential effects on motor behavior and sensorimotor network function. Exp Neurol 2023; 367:114471. [PMID: 37321386 PMCID: PMC10695146 DOI: 10.1016/j.expneurol.2023.114471] [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: 02/17/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
Dystonia is a neurological movement disorder characterized by repetitive, unintentional movements and disabling postures that result from sustained or intermittent muscle contractions. The basal ganglia and cerebellum have received substantial focus in studying DYT1 dystonia. It remains unclear how cell-specific ∆GAG mutation of torsinA within specific cells of the basal ganglia or cerebellum affects motor performance, somatosensory network connectivity, and microstructure. In order to achieve this goal, we generated two genetically modified mouse models: in model 1 we performed Dyt1 ∆GAG conditional knock-in (KI) in neurons that express dopamine-2 receptors (D2-KI), and in model 2 we performed Dyt1 ∆GAG conditional KI in Purkinje cells of the cerebellum (Pcp2-KI). In both of these models, we used functional magnetic resonance imaging (fMRI) to assess sensory-evoked brain activation and resting-state functional connectivity, and diffusion MRI to assess brain microstructure. We found that D2-KI mutant mice had motor deficits, abnormal sensory-evoked brain activation in the somatosensory cortex, as well as increased functional connectivity of the anterior medulla with cortex. In contrast, we found that Pcp2-KI mice had improved motor performance, reduced sensory-evoked brain activation in the striatum and midbrain, as well as reduced functional connectivity of the striatum with the anterior medulla. These findings suggest that (1) D2 cell-specific Dyt1 ∆GAG mediated torsinA dysfunction in the basal ganglia results in detrimental effects on the sensorimotor network and motor output, and (2) Purkinje cell-specific Dyt1 ∆GAG mediated torsinA dysfunction in the cerebellum results in compensatory changes in the sensorimotor network that protect against dystonia-like motor deficits.
Collapse
Affiliation(s)
- B J Wilkes
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA.
| | - R Z Adury
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - D Berryman
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | - L R Concepcion
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Y Liu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | - F Yokoi
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | - C Maugee
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Y Li
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA
| | - D E Vaillancourt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Norman Fixel Institute for Neurological Diseases, Department of Neurology, University of Florida, Gainesville, FL, USA; Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
7
|
Stephen CD, Dy-Hollins M, Gusmao CMD, Qahtani XA, Sharma N. Dystonias: Clinical Recognition and the Role of Additional Diagnostic Testing. Semin Neurol 2023; 43:17-34. [PMID: 36972613 DOI: 10.1055/s-0043-1764292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Dystonia is the third most common movement disorder, characterized by abnormal, frequently twisting postures related to co-contraction of agonist and antagonist muscles. Diagnosis is challenging. We provide a comprehensive appraisal of the epidemiology and an approach to the phenomenology and classification of dystonia, based on the clinical characteristics and underlying etiology of dystonia syndromes. We discuss the features of common idiopathic and genetic forms of dystonia, diagnostic challenges, and dystonia mimics. Appropriate workup is based on the age of symptom onset, rate of progression, whether dystonia is isolated or combined with another movement disorder or complex neurological and other organ system eatures. Based on these features, we discuss when imaging and genetic should be considered. We discuss the multidisciplinary treatment of dystonia, including rehabilitation and treatment principles according to the etiology, including when pathogenesis-direct treatment is available, oral pharmacological therapy, chemodenervation with botulinum toxin injections, deep brain stimulation and other surgical therapies, and future directions.
Collapse
Affiliation(s)
| | - Marisela Dy-Hollins
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Xena Al Qahtani
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
| | - Nutan Sharma
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
8
|
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
|
9
|
Rafee S, Al-Hinai M, Hutchinson M. Adult-Onset Idiopathic Cervical Dystonia. EUROPEAN MEDICAL JOURNAL 2022. [DOI: 10.33590/emj/10005730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Adult-onset idiopathic focal dystonia is the most common type of primary dystonia, and adult-onset idiopathic cervical dystonia (AOICD) is its most prevalent phenotype. AOICD is an autosomal-dominant disorder with markedly reduced penetrance; clinical expression is dependent on age, sex, and environmental exposure. Motor symptoms at presentation are poorly recognised by non-specialists, leading to long delays in diagnosis. Certain features of history and examination can help diagnose cervical dystonia. There is a relatively high prevalence of anxiety and/or depression, which adversely affects health-related quality of life. Recent studies indicate that patients with AOICD also have disordered social cognition, particularly affecting emotional sensory processing. AOICD can be treated reasonably effectively with botulinum toxin injections, given at 3-month intervals. Oral antidystonic medications are often trialled initially, but are largely ineffective. Comprehensive modern management of patients with AOICD requires recognition of presence of mood disorders, and actively treating the endogenous mood disorder with antidepressant therapy. Botulinum toxin injections alone, no matter how expertly given, will not provide optimal therapy and improved health-related quality of life without an holistic approach to patient management. Increasing evidence indicates that AOICD is a neurophysiological network disorder of GABAergic inhibition, causing a syndrome of dystonia, mood disturbance, and social cognitive dysfunction, with the superior colliculus playing a central role.
Collapse
Affiliation(s)
- Shameer Rafee
- Department of Neurology, St Vincent’s University Hospital, Dublin, Republic of Ireland
| | - Mahmood Al-Hinai
- Department of Neurology, St Vincent’s University Hospital, Dublin, Republic of Ireland
| | - Michael Hutchinson
- Department of Neurology, St Vincent’s University Hospital, Dublin, Republic of Ireland
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW This article discusses the most recent findings regarding the diagnosis, classification, and management of genetic and idiopathic dystonia. RECENT FINDINGS A new approach to classifying dystonia has been created with the aim to increase the recognition and diagnosis of dystonia. Molecular biology and genetic studies have identified several genes and biological pathways involved in dystonia. SUMMARY Dystonia is a common movement disorder involving abnormal, often twisting, postures and is a challenging condition to diagnose. The pathophysiology of dystonia involves abnormalities in brain motor networks in the context of genetic factors. Dystonia has genetic, idiopathic, and acquired forms, with a wide phenotypic spectrum, and is a common feature in complex neurologic disorders. Dystonia can be isolated or combined with another movement disorder and may be focal, segmental, multifocal, or generalized in distribution, with some forms only occurring during the performance of specific tasks (task-specific dystonia). Dystonia is classified by clinical characteristics and presumed etiology. The management of dystonia involves accurate diagnosis, followed by treatment with botulinum toxin injections, oral medications, and surgical therapies (mainly deep brain stimulation), as well as pathogenesis-directed treatments, including the prospect of disease-modifying or gene therapies.
Collapse
|
11
|
Downs AM, Donsante Y, Jinnah H, Hess EJ. Blockade of M4 muscarinic receptors on striatal cholinergic interneurons normalizes striatal dopamine release in a mouse model of TOR1A dystonia. Neurobiol Dis 2022; 168:105699. [DOI: 10.1016/j.nbd.2022.105699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/10/2022] [Accepted: 03/15/2022] [Indexed: 10/18/2022] Open
|
12
|
Aryal R, Shrestha S, Homagain S, Chhetri S, Shrestha K, Kharel S, Karn R, Rajbhandari R, Gajurel BP, Ojha R. Clinical spectrum and management of dystonia in patients with Japanese encephalitis: A systematic review. Brain Behav 2022; 12:e2496. [PMID: 35025122 PMCID: PMC8865161 DOI: 10.1002/brb3.2496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/25/2021] [Accepted: 01/02/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Japanese encephalitis (JE) is a potentially fatal viral infection with a wide range of manifestations and can also present with a variety of movement disorders (MD) including dystonia. Dystonic features in JE are uncommon. Here, we have tried to summarize the clinical features and management of dystonia among JE patients with a comprehensive literature search. METHODS Various databases, including PubMed, Embase, and Google Scholar, were searched against the predefined criteria using suitable keywords combination and boolean operations. Relevant information from observational and case studies was extracted according to the author, dystonic features, radiological changes in the brain scans, treatment options, and outcome wherever provided. RESULT We identified 19 studies with a total of 1547 JE patients, the diagnosis of which was confirmed by IgM detection in serum and/or cerebrospinal fluid in the majority of the patients (88.62%). 234 (15.13%) of JE patients had dystonia with several types of focal dystonia being present in 131 (55.98%) either alone or in combination. Neuroimaging showed predominant involvement of thalami, basal ganglia, and brainstem. Oral medications including anticholinergics, GABA agonists, and benzodiazepines followed by botulinum toxin were the most common treatment modalities. CONCLUSION Dystonia can be a disabling consequence of JE, and various available medical therapies can significantly improve the quality of life. Owing to insufficient studies on the assessment of dystonia associated with JE, longitudinal studies with a larger number of patients are warranted to further clarify the clinical course, treatment, and outcome of dystonia.
Collapse
Affiliation(s)
- Roshan Aryal
- Department of Medicine, Maharajgunj Medical CampusInstitute of MedicineKathmandu44600Nepal
| | - Suraj Shrestha
- Department of Medicine, Maharajgunj Medical CampusInstitute of MedicineKathmandu44600Nepal
| | - Sushan Homagain
- Department of Medicine, Maharajgunj Medical CampusInstitute of MedicineKathmandu44600Nepal
| | - Sunit Chhetri
- Department of MedicineB.P. Koirala Institute of Health SciencesDharan56700Nepal
| | - Kshitiz Shrestha
- Department of MedicineB.P. Koirala Institute of Health SciencesDharan56700Nepal
| | - Sanjeev Kharel
- Department of Medicine, Maharajgunj Medical CampusInstitute of MedicineKathmandu44600Nepal
| | - Ragesh Karn
- Department of NeurologyTribhuvan University Institute of MedicineKathmandu44600Nepal
| | - Reema Rajbhandari
- Department of NeurologyTribhuvan University Institute of MedicineKathmandu44600Nepal
| | - Bikram Prasad Gajurel
- Department of NeurologyTribhuvan University Institute of MedicineKathmandu44600Nepal
| | - Rajeev Ojha
- Department of NeurologyTribhuvan University Institute of MedicineKathmandu44600Nepal
| |
Collapse
|
13
|
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: 1.0] [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
|
14
|
Moehle MS, Bender AM, Dickerson JW, Foster DJ, Qi A, Cho HP, Donsante Y, Peng W, Bryant Z, Stillwell KJ, Bridges TM, Chang S, Watson KJ, O’Neill JC, Engers JL, Peng L, Rodriguez AL, Niswender CM, Lindsley CW, Hess EJ, Conn PJ, Rook JM. Discovery of the First Selective M 4 Muscarinic Acetylcholine Receptor Antagonists with in Vivo Antiparkinsonian and Antidystonic Efficacy. ACS Pharmacol Transl Sci 2021; 4:1306-1321. [PMID: 34423268 PMCID: PMC8369681 DOI: 10.1021/acsptsci.0c00162] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Indexed: 11/30/2022]
Abstract
Nonselective antagonists of muscarinic acetylcholine receptors (mAChRs) that broadly inhibit all five mAChR subtypes provide an efficacious treatment for some movement disorders, including Parkinson's disease and dystonia. Despite their efficacy in these and other central nervous system disorders, antimuscarinic therapy has limited utility due to severe adverse effects that often limit their tolerability by patients. Recent advances in understanding the roles that each mAChR subtype plays in disease pathology suggest that highly selective ligands for individual subtypes may underlie the antiparkinsonian and antidystonic efficacy observed with the use of nonselective antimuscarinic therapeutics. Our recent work has indicated that the M4 muscarinic acetylcholine receptor has several important roles in opposing aberrant neurotransmitter release, intracellular signaling pathways, and brain circuits associated with movement disorders. This raises the possibility that selective antagonists of M4 may recapitulate the efficacy of nonselective antimuscarinic therapeutics and may decrease or eliminate the adverse effects associated with these drugs. However, this has not been directly tested due to lack of selective antagonists of M4. Here, we utilize genetic mAChR knockout animals in combination with nonselective mAChR antagonists to confirm that the M4 receptor activation is required for the locomotor-stimulating and antiparkinsonian efficacy in rodent models. We also report the synthesis, discovery, and characterization of the first-in-class selective M4 antagonists VU6013720, VU6021302, and VU6021625 and confirm that these optimized compounds have antiparkinsonian and antidystonic efficacy in pharmacological and genetic models of movement disorders.
Collapse
Affiliation(s)
- Mark S. Moehle
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Department
of Pharmacology & Therapeutics, Center for Translational Research
in Neurodegeneration, University of Florida, Gainesville, Florida 32610, United States
| | - Aaron M. Bender
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jonathan W. Dickerson
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Daniel J. Foster
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Aidong Qi
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Hyekyung P. Cho
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Yuping Donsante
- Department
of Pharmacology & Chemical Biology, Emory University, Atlanta, Georgia 30322, United States
| | - Weimin Peng
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Zoey Bryant
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Kaylee J. Stillwell
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Sichen Chang
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Katherine J. Watson
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jordan C. O’Neill
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Julie L. Engers
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Li Peng
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Alice L. Rodriguez
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Colleen M. Niswender
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Ellen J. Hess
- Department
of Pharmacology & Chemical Biology, Emory University, Atlanta, Georgia 30322, United States
| | - P. Jeffrey Conn
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,Vanderbilt
Kennedy Center, Vanderbilt University, Nashville, Tennessee 37232, United States,E-mail:
| | - Jerri M. Rook
- Department
of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States,E-mail:
| |
Collapse
|
15
|
Svetel M, Tomić A, Kresojević N, Dragašević N, Kostić V. Perspectives on the pharmacological management of dystonia. Expert Opin Pharmacother 2021; 22:1555-1566. [PMID: 33904811 DOI: 10.1080/14656566.2021.1919083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Treatment of dystonia is particularly complex due to various etiologies and heterogeneous clinical manifestation, as well as different degrees of disability. In absence of causative treatment, all symptomatic therapy should be predominantly tailored to ameliorate those symptoms (motor and non/motor) that mostly affect patients' daily life and regular activities. Many different treatment options, including oral medications, neurosurgical interventions, physical and occupational therapy are available in treatment of dystonia.Areas covered: The aim of this perspective is to point out different possibilities in pharmacological management of dystonic movements. Due to pure clinical presentation, the authors concentrate mainly on the isolated dystonias, which are presented solely as dystonic movements. Combined and complex dystonias are not instructive due to compound clinical presentation and consequently, complicated treatment. The article is based on a literature search from sources including PubMed, the Cochrane Library, Web of Science, PiCarta, and PsycINFO.Expert opinion: Although dystonia therapy should be adapted according to the individual needs, severity, age, type, symptoms distribution and acceptable side-effect profile, certain principles should be followed to reach the optimal result. Furthermore, the authors believe that a better understanding of the pathophysiology of dystonia will bring with it the development of new and improved treatment approaches and medications.
Collapse
Affiliation(s)
- Marina Svetel
- Movement Disorders Department, Clinic of Neurology, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Tomić
- Movement Disorders Department, Clinic of Neurology, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nikola Kresojević
- Movement Disorders Department, Clinic of Neurology, Clinical Center of Serbia, Belgrade, Serbia
| | - Nataša Dragašević
- Movement Disorders Department, Clinic of Neurology, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir Kostić
- Movement Disorders Department, Clinic of Neurology, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
16
|
Downs AM, Fan X, Kadakia RF, Donsante Y, Jinnah HA, Hess EJ. Cell-intrinsic effects of TorsinA(ΔE) disrupt dopamine release in a mouse model of TOR1A dystonia. Neurobiol Dis 2021; 155:105369. [PMID: 33894367 DOI: 10.1016/j.nbd.2021.105369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 11/19/2022] Open
Abstract
TOR1A-associated dystonia, otherwise known as DYT1 dystonia, is an inherited dystonia caused by a three base-pair deletion in the TOR1A gene (TOR1AΔE). Although the mechanisms underlying the dystonic movements are largely unknown, abnormalities in striatal dopamine and acetylcholine neurotransmission are consistently implicated whereby dopamine release is reduced while cholinergic tone is increased. Because striatal cholinergic neurotransmission mediates dopamine release, it is not known if the dopamine release deficit is mediated indirectly by abnormal acetylcholine neurotransmission or if Tor1a(ΔE) acts directly within dopaminergic neurons to attenuate release. To dissect the microcircuit that governs the deficit in dopamine release, we conditionally expressed Tor1a(ΔE) in either dopamine neurons or cholinergic interneurons in mice and assessed striatal dopamine release using ex vivo fast scan cyclic voltammetry or dopamine efflux using in vivo microdialysis. Conditional expression of Tor1a(ΔE) in cholinergic neurons did not affect striatal dopamine release. In contrast, conditional expression of Tor1a(ΔE) in dopamine neurons reduced dopamine release to 50% of normal, which is comparable to the deficit in Tor1a+/ΔE knockin mice that express the mutation ubiquitously. Despite the deficit in dopamine release, we found that the Tor1a(ΔE) mutation does not cause obvious nerve terminal dysfunction as other presynaptic mechanisms, including electrical excitability, vesicle recycling/refilling, Ca2+ signaling, D2 dopamine autoreceptor function and GABAB receptor function, are intact. Although the mechanistic link between Tor1a(ΔE) and dopamine release is unclear, these results clearly demonstrate that the defect in dopamine release is caused by the action of the Tor1a(ΔE) mutation within dopamine neurons.
Collapse
Affiliation(s)
- Anthony M Downs
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - Xueliang Fan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - Radhika F Kadakia
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - Yuping Donsante
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - H A Jinnah
- Department of Neurology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA; Department of Human Genetics, Emory University School of Medicine, 101 Woodruff Circle, WMB 6300, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, 101 Woodruff Circle, WMB 6300, Atlanta, GA 30322, USA
| | - Ellen J Hess
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA; Department of Neurology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA.
| |
Collapse
|
17
|
Dystonia and leveraging oral pharmacotherapy. J Neural Transm (Vienna) 2021; 128:521-529. [PMID: 33877451 DOI: 10.1007/s00702-021-02339-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 04/15/2021] [Indexed: 11/27/2022]
Abstract
Dystonia is a clinically diverse disorder, characterized by sustained or intermittent muscle contractions causing abnormal and often repetitive movements and/or postures. Accurate clinical diagnosis is tantamount to effective dystonia management. Current guidelines in the treatments of dystonia, including oral therapy, are prescribed to improve symptoms and to restore functional capacity. Identifying treatable causes from co-existing phenomenologies is relevant to provide the most optimal and disease-specific medications. In other forms of dystonia, genetic factors may affect outcome. Moreover, proper selection of patients, early initiation of medications and customized drug titration are keys to increasing the chances of success when using oral therapies for dystonia. Treatment of dystonia primarily involves agents that target dopamine and acetylcholine receptors. Other drugs used include benzodiazepines, baclofen, antiepileptics, some antipsychotics drugs and antihistamine, with different levels of evidence of effectiveness. Unfortunately, most of the widely used drugs have low levels of evidence and are primarily based on anecdotal experiences. Finally, other adjunctive therapeutic strategies are often necessary to complement oral therapy.
Collapse
|
18
|
Okamoto N, Konishi Y, Tesen H, Ikenouchi A, Yoshimura R. A Low Clozapine Dose Improved Refractory Tardive Dystonia without Exacerbating Psychiatric Symptoms: A Case Report. Int Med Case Rep J 2021; 14:237-239. [PMID: 33889030 PMCID: PMC8057797 DOI: 10.2147/imcrj.s307410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/07/2021] [Indexed: 01/19/2023] Open
Abstract
Clozapine is recommended for patients with schizophrenia and tardive dystonia (TD); however, the appropriate dose remains unclear. In this case, a low dose (150 mg/day) of clozapine improved refractory TD and further ameliorated psychiatric symptoms. Herein, we report on a 41-year-old female with schizophrenia and TD who was treated with a low clozapine dose. After eight weeks of continuous clozapine at 150 mg/day (16 weeks after clozapine initiation), her TD dramatically improved, and her psychiatric symptoms were relieved. Low clozapine doses could ameliorate refractory TD. However, this effect might require up to several weeks. Clinicians should be patient unless they consider it better to increase the clozapine dose.
Collapse
Affiliation(s)
- Naomichi Okamoto
- Department of Psychiatry, University of Occupational and Environmental Health Kitakyushu, Fukuoka, 8078555, Japan
| | - Yuki Konishi
- Department of Psychiatry, University of Occupational and Environmental Health Kitakyushu, Fukuoka, 8078555, Japan
| | - Hirofumi Tesen
- Department of Psychiatry, University of Occupational and Environmental Health Kitakyushu, Fukuoka, 8078555, Japan
| | - Atsuko Ikenouchi
- Department of Psychiatry, University of Occupational and Environmental Health Kitakyushu, Fukuoka, 8078555, Japan.,Medical Center for Dementia, University Hospital, University of Occupational and Environmental Health Kitakyushu, Fukuoka, 8078555, Japan
| | - Reiji Yoshimura
- Department of Psychiatry, University of Occupational and Environmental Health Kitakyushu, Fukuoka, 8078555, Japan
| |
Collapse
|
19
|
Complex dystonias: an update on diagnosis and care. J Neural Transm (Vienna) 2020; 128:431-445. [PMID: 33185802 PMCID: PMC8099829 DOI: 10.1007/s00702-020-02275-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023]
Abstract
Complex dystonias are defined as dystonias that are accompanied by neurologic or systemic manifestations beyond movement disorders. Many syndromes or diseases can present with complex dystonia, either as the cardinal sign or as part of a multi-systemic manifestation. Complex dystonia often gradually develops in the disease course, but can also be present from the outset. If available, the diagnostic workup, disease-specific treatment, and management of patients with complex dystonias require a multi-disciplinary approach. This article summarizes current knowledge on complex dystonias with a particular view of recent developments with respect to advances in diagnosis and management, including causative treatments.
Collapse
|
20
|
Bledsoe IO, Viser AC, San Luciano M. Treatment of Dystonia: Medications, Neurotoxins, Neuromodulation, and Rehabilitation. Neurotherapeutics 2020; 17:1622-1644. [PMID: 33095402 PMCID: PMC7851280 DOI: 10.1007/s13311-020-00944-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2020] [Indexed: 02/24/2023] Open
Abstract
Dystonia is a complex disorder with numerous presentations occurring in isolation or in combination with other neurologic symptoms. Its treatment has been significantly improved with the advent of botulinum toxin and deep brain stimulation in recent years, though additional investigation is needed to further refine these interventions. Medications are of critical importance in forms of dopa-responsive dystonia but can be beneficial in other forms of dystonia as well. Many different rehabilitative paradigms have been studied with variable benefit. There is growing interest in noninvasive stimulation as a potential treatment, but with limited long-term benefit shown to date, and additional research is needed. This article reviews existing evidence for treatments from each of these categories. To date, there are many examples of incomplete response to available treatments, and improved therapies are needed.
Collapse
Affiliation(s)
- Ian O. Bledsoe
- Weill Institute for Neurosciences, Movement Disorder and Neuromodulation Center, University of California, San Francisco, 1635 Divisadero St., Suite 520, San Francisco, CA 94115 USA
| | - Aaron C. Viser
- Weill Institute for Neurosciences, Movement Disorder and Neuromodulation Center, University of California, San Francisco, 1635 Divisadero St., Suite 520, San Francisco, CA 94115 USA
| | - Marta San Luciano
- Weill Institute for Neurosciences, Movement Disorder and Neuromodulation Center, University of California, San Francisco, 1635 Divisadero St., Suite 520, San Francisco, CA 94115 USA
| |
Collapse
|
21
|
|
22
|
Long-Term Response to Clozapine and Its Clinical Correlates in the Treatment of Tardive Movement Syndromes: A Naturalistic Observational Study in Patients With Psychotic Disorders. J Clin Psychopharmacol 2020; 39:591-596. [PMID: 31688397 DOI: 10.1097/jcp.0000000000001114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Given that switching to clozapine is an important treatment option for tardive movement syndrome (TMS), its effect and clinical correlates have not been fully explored yet. This study investigated the improvement of TMS after switching to clozapine and factors associated with the response in a naturalistic outpatient setting. METHODS Subjects were 35 patients with schizophrenia or bipolar disorder receiving only clozapine as an antipsychotic drug for more than 12 months. Their prior antipsychotics were switched to clozapine after the onset of tardive dyskinesia and/or tardive dystonia. Tardive movement syndrome and clinical characteristics were assessed through direct examination and review of hospital records. FINDINGS Offending antipsychotics administered at the time of TMS onset were second-generation antipsychotics in 88.6% of patients. Tardive movement syndrome symptoms were remitted in 65.7% of patients after switching to clozapine. Younger age, younger age at onset of TMS, and lower baseline Abnormal Involuntary Movement Scale score were significantly associated with remission of TMS. Female sex and good antipsychotic effects of clozapine showed a trend of association with better response. IMPLICATIONS Clozapine seems to be an excellent treatment option for TMS in the era of second-generation antipsychotics, especially for younger patients with mild tardive dyskinesia. Clinical trials comparing the effect of switching antipsychotics to clozapine with add-on therapy of new drugs targeting TMS are difficult to design in ordinary clinical settings. Therefore, more naturalistic observational studies are warranted to identify predictors of TMS response to clozapine.
Collapse
|
23
|
Wojtasiewicz T, Butala A, Anderson WS. Dystonia. Stereotact Funct Neurosurg 2020. [DOI: 10.1007/978-3-030-34906-6_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
24
|
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: 1.0] [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]
|
25
|
Bellows S, Jankovic J. Treatment of dystonia and tics. Clin Park Relat Disord 2019; 2:12-19. [PMID: 34316614 PMCID: PMC8302199 DOI: 10.1016/j.prdoa.2019.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 11/30/2022] Open
Abstract
Treatment of dystonia and tics continues to evolve. In dystonia, while oral agents such as benzodiazepines, baclofen and anticholinergics remain in use, botulinum toxin (BoNT) continues to be regarded as the treatment of choice for focal and segmental dystonia, but new preparations are being studied. While deep brain stimulation (DBS) has typically focused on targeting the globus pallidus internus (GPi) when treating dystonia, more recent research has expanded the targets to include subthalamic nucleus (STN) and other targets. In addition to DBS, thalamotomies continue to show therapeutic benefit in focal hand dystonias. Treatment of tics includes a growing armamentarium of options besides the three FDA-approved drugs, all dopamine receptor blockers (haloperidol, pimozide and aripiprazole). Because of lower risk of adverse effects, dopamine depleters (e.g. tetrabebazine, deutetrabenazine, and valbenazine), along with novel D1 receptor antagonists, are currently studied as treatment alternatives in patients with tics. Practice guidelines for the treatment of tics and Tourette syndrome have been recently updated. Data regarding the use of DBS in treatment of tics remains relatively sparse, but international registries have expanded our understanding of the effect of stimulation at several targets.
Collapse
Affiliation(s)
- Steven Bellows
- Parkinson's Disease Center, Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Joseph Jankovic
- Parkinson's Disease Center, Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
26
|
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: 30] [Impact Index Per Article: 5.0] [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
|
27
|
Da Silva JD, Teixeira-Castro A, Maciel P. From Pathogenesis to Novel Therapeutics for Spinocerebellar Ataxia Type 3: Evading Potholes on the Way to Translation. Neurotherapeutics 2019; 16:1009-1031. [PMID: 31691128 PMCID: PMC6985322 DOI: 10.1007/s13311-019-00798-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease (MJD), is a neurodegenerative disorder caused by a polyglutamine expansion in the ATXN3 gene. In spite of the identification of a clear monogenic cause 25 years ago, the pathological process still puzzles researchers, impairing prospects for an effective therapy. Here, we propose the disruption of protein homeostasis as the hub of SCA3 pathogenesis, being the molecular mechanisms and cellular pathways that are deregulated in SCA3 downstream consequences of the misfolding and aggregation of ATXN3. Moreover, we attempt to provide a realistic perspective on how the translational/clinical research in SCA3 should evolve. This was based on molecular findings, clinical and epidemiological characteristics, studies of proposed treatments in other conditions, and how that information is essential for their (re-)application in SCA3. This review thus aims i) to critically evaluate the current state of research on SCA3, from fundamental to translational and clinical perspectives; ii) to bring up the current key questions that remain unanswered in this disorder; and iii) to provide a frame on how those answers should be pursued.
Collapse
Affiliation(s)
- Jorge Diogo Da Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
28
|
Hirjak D, Kubera KM, Bienentreu S, Thomann PA, Wolf RC. [Antipsychotic-induced motor symptoms in schizophrenic psychoses-Part 1 : Dystonia, akathisia und parkinsonism]. DER NERVENARZT 2019; 90:1-11. [PMID: 30128734 DOI: 10.1007/s00115-018-0582-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Acute antipsychotic-induced movement disorders (AIMD) are clinically relevant since they are frequently associated with high subjective distress, and since over the long-term they can negatively impact treatment adherence of patients with schizophrenic psychoses. This review article summarizes the relevant studies on the prevalence, risk factors, prevention and treatment options and instruments for early prediction of acute AIMD in schizophrenic psychoses. The current evidence and treatment recommendations are divided into three main areas: acute dystonia, akathisia, and parkinsonism. For the treatment of acute dystonia trihexyphenidyl and biperiden have shown their efficacy. Considering pharmacological treatment of akathisia, there is some preliminary evidence for medication with lipophilic beta-receptor blockers (propranolol and pindolol), clonidine, benzodiazepines, mianserin, mirtazapine und trazodone. The treatment options for drug-induced parkinsonism include reduction or switching from one antipsychotic to another with a lower affinity for dopamine D2 receptors, amantadine or in the regular administration of anticholinergic drugs. In conclusion, acute AIMD is easily to recognize but is not always effectively and durably treated. Early recognition and treatment of acute AIMD could be associated with improved treatment outcomes.
Collapse
Affiliation(s)
- D Hirjak
- Zentralinstitut für Seelische Gesundheit, Klinik für Psychiatrie und Psychotherapie, Medizinische Fakultät Mannheim, Universität Heidelberg, J5, 68159, Mannheim, Deutschland.
| | - K M Kubera
- Zentrum für Psychosoziale Medizin, Klinik für Allgemeine Psychiatrie, Universität Heidelberg, Heidelberg, Deutschland
| | - S Bienentreu
- Fachklinik für Psychiatrie und Psychotherapie, MARIENBORN GmbH, Zülpich, Deutschland
| | - P A Thomann
- Zentrum für Psychosoziale Medizin, Klinik für Allgemeine Psychiatrie, Universität Heidelberg, Heidelberg, Deutschland
- Zentrum für Seelische Gesundheit, Gesundheitszentrum Odenwaldkreis, Erbach, Deutschland
| | - R C Wolf
- Zentrum für Psychosoziale Medizin, Klinik für Allgemeine Psychiatrie, Universität Heidelberg, Heidelberg, Deutschland
| |
Collapse
|
29
|
Diverse Mechanisms Lead to Common Dysfunction of Striatal Cholinergic Interneurons in Distinct Genetic Mouse Models of Dystonia. J Neurosci 2019; 39:7195-7205. [PMID: 31320448 DOI: 10.1523/jneurosci.0407-19.2019] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/16/2022] Open
Abstract
Clinical and experimental data indicate striatal cholinergic dysfunction in dystonia, a movement disorder typically resulting in twisted postures via abnormal muscle contraction. Three forms of isolated human dystonia result from mutations in the TOR1A (DYT1), THAP1 (DYT6), and GNAL (DYT25) genes. Experimental models carrying these mutations facilitate identification of possible shared cellular mechanisms. Recently, we reported elevated extracellular striatal acetylcholine by in vivo microdialysis and paradoxical excitation of cholinergic interneurons (ChIs) by dopamine D2 receptor (D2R) agonism using ex vivo slice electrophysiology in Dyt1 ΔGAG/+ mice. The paradoxical excitation was caused by overactive muscarinic receptors (mAChRs), leading to a switch in D2R coupling from canonical Gi/o to noncanonical β-arrestin signaling. We sought to determine whether these mechanisms in Dyt1 ΔGAG/+ mice are shared with Thap1 C54Y/+ knock-in and Gnal +/- knock-out dystonia models and to determine the impact of sex. We found Thap1 C54Y/+ mice of both sexes have elevated extracellular striatal acetylcholine and D2R-induced paradoxical ChI excitation, which was reversed by mAChR inhibition. Elevated extracellular acetylcholine was absent in male and female Gnal +/- mice, but the paradoxical D2R-mediated ChI excitation was retained and only reversed by inhibition of adenosine A2ARs. The Gi/o-preferring D2R agonist failed to increase ChI excitability, suggesting a possible switch in coupling of D2Rs to β-arrestin, as seen previously in a DYT1 model. These data show that, whereas elevated extracellular acetylcholine levels are not always detected across these genetic models of human dystonia, the D2R-mediated paradoxical excitation of ChIs is shared and is caused by altered function of distinct G-protein-coupled receptors.SIGNIFICANCE STATEMENT Dystonia is a common and often disabling movement disorder. The usual medical treatment of dystonia is pharmacotherapy with nonselective antagonists of muscarinic acetylcholine receptors, which have many undesirable side effects. Development of new therapeutics is a top priority for dystonia research. The current findings, considered in context with our previous investigations, establish a role for cholinergic dysfunction across three mouse models of human genetic dystonia: DYT1, DYT6, and DYT25. The commonality of cholinergic dysfunction in these models arising from diverse molecular etiologies points the way to new approaches for cholinergic modulation that may be broadly applicable in dystonia.
Collapse
|
30
|
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: 24] [Impact Index Per Article: 4.0] [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
|
31
|
Pirio Richardson S, Jinnah HA. New approaches to discovering drugs that treat dystonia. Expert Opin Drug Discov 2019; 14:893-900. [PMID: 31159587 DOI: 10.1080/17460441.2019.1623785] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Dystonia consists of involuntary movements, abnormal posturing, and pain. In adults, dystonia presents in a particular region of the body and causes significant disability due to pain as well as impairment in activities of daily living and employment. The current gold standard treatment, botulinum toxin (BoNT), has limitations - painful, frequent injections due to 'wearing off' of treatment effect; expense; and expected side effects like swallowing difficulty and weakness. There is a clear therapeutic gap in our current treatment options for dystonia and also a clear need for an effective novel treatment. Testing any novel treatment is complicated because most adults with focal dystonia are treated with BoNT. Areas covered: This review focuses on establishing the need for novel therapeutics. It also suggests potential leads from preclinical studies; and, discusses the issue of clinical trial readiness in the dystonia field. Expert opinion: Identifying a novel therapeutic intervention for dystonia patients faces two major challenges. The first is acknowledging the therapeutic gap that currently exists. Second, shifting some of our research aims in dystonia to clinical trial readiness is imperative if we are to be ready to test novel therapeutic agents.
Collapse
Affiliation(s)
- Sarah Pirio Richardson
- a Department of Neurology, University of New Mexico Health Sciences Center , Albuquerque , NM , USA.,b Neurology Service, New Mexico Veterans Affairs Health Care System , Albuquerque , NM , USA
| | - H A Jinnah
- c Departments of Neurology, Human Genetics & Pediatrics, Emory University School of Medicine , Atlanta , Georgia
| |
Collapse
|
32
|
Mohammad SS, Paget SP, Dale RC. Current therapies and therapeutic decision making for childhood-onset movement disorders. Mov Disord 2019; 34:637-656. [PMID: 30919519 DOI: 10.1002/mds.27661] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 02/12/2019] [Indexed: 12/13/2022] Open
Abstract
Movement disorders differ in children to adults. First, neurodevelopmental movement disorders such as tics and stereotypies are more prevalent than parkinsonism, and second, there is a genomic revolution which is now explaining many early-onset dystonic syndromes. We outline an approach to children with movement disorders starting with defining the movement phenomenology, determining the level of functional impairment due to abnormal movements, and screening for comorbid psychiatric conditions and cognitive impairments which often contribute more to disability than the movements themselves. The rapid improvement in our understanding of the etiology of movement disorders has resulted in an increasing focus on precision medicine, targeting treatable conditions and defining modifiable disease processes. We profile some of the key disease-modifying therapies in metabolic, neurotransmitter, inflammatory, and autoimmune conditions and the increasing focus on gene or cellular therapies. When no disease-modifying therapies are possible, symptomatic therapies are often all that is available. These classically target dopaminergic, cholinergic, alpha-adrenergic, or GABAergic neurochemistry. Increasing interest in neuromodulation has highlighted that some clinical syndromes respond better to DBS, and further highlights the importance of "disease-specific" therapies with a future focus on individualized therapies according to the genomic findings or disease pathways that are disrupted. We summarize some pragmatic applications of symptomatic therapies, neuromodulation techniques, and some rehabilitative interventions and provide a contemporary overview of treatment in childhood-onset movement disorders. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Shekeeb S Mohammad
- Kids Neuroscience Centre, The Kids Research Institute at the Children's Hospital at Westmead, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia.,Movement Disorders Unit, T.Y. Nelson Department of Neurology, the Children's Hospital at Westmead and Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Simon P Paget
- Kids Rehab, the Children's Hospital at Westmead and Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Russell C Dale
- Kids Neuroscience Centre, The Kids Research Institute at the Children's Hospital at Westmead, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia.,Movement Disorders Unit, T.Y. Nelson Department of Neurology, the Children's Hospital at Westmead and Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
33
|
Dietz N, Neimat J. Neuromodulation: Deep Brain Stimulation for Treatment of Dystonia. Neurosurg Clin N Am 2019; 30:161-168. [PMID: 30898268 DOI: 10.1016/j.nec.2018.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dystonia is a heterogeneous, hyperkinetic movement disorder with sustained or intermittent abnormal postures, hyperkinetic muscle contractions, or repetitive movements. Classification of dystonia involves 2 axes: axis I and axis II, defining relevant clinical features and etiology, respectively. Medical therapy varies based on subtype and includes intramuscular botulinum toxin injections and oral anticholinergic pharmaceuticals. Deep brain stimulation became widely incorporated in 1999 after several landmark studies and has been effectively used in targets of the thalamus, pallidum, and subthalamic nucleus. New insights into pathophysiology of dystonia and genetic analysis continue to guide surgical technique toward ever-effective treatment.
Collapse
Affiliation(s)
- Nicholas Dietz
- Department of Neurosurgery, University of Louisville, School of Medicine, 200 Abraham Flexner Highway, Louisville, KY 40202, USA
| | - Joseph Neimat
- Department of Neurosurgery, University of Louisville, School of Medicine, 200 Abraham Flexner Highway, Louisville, KY 40202, USA.
| |
Collapse
|
34
|
Downs AM, Fan X, Donsante C, Jinnah HA, Hess EJ. Trihexyphenidyl rescues the deficit in dopamine neurotransmission in a mouse model of DYT1 dystonia. Neurobiol Dis 2019; 125:115-122. [PMID: 30707939 DOI: 10.1016/j.nbd.2019.01.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/14/2019] [Accepted: 01/20/2019] [Indexed: 11/17/2022] Open
Abstract
Trihexyphenidyl, a nonselective muscarinic receptor antagonist, is the small molecule drug of choice for the treatment of DYT1 dystonia, but it is poorly tolerated due to significant side effects. A better understanding of the mechanism of action of trihexyphenidyl is needed for the development of improved treatments. Because DTY1 dystonia is associated with both abnormal cholinergic neurotransmission and abnormal dopamine regulation, we tested the hypothesis that trihexyphenidyl normalizes striatal dopamine release in a mouse model of DYT1 dystonia using ex vivo fast scan cyclic voltammetry and in vivo microdialysis. Trihexyphenidyl increased striatal dopamine release and efflux as assessed by ex vivo voltammetry and in vivo microdialysis respectively. In contrast, ʟ-DOPA, which is not usually effective for the treatment of DYT1 dystonia, did not increase dopamine release in either Dyt1 or control mice. Trihexyphenidyl was less effective at enhancing dopamine release in Dyt1 mice relative to controls ex vivo (mean increase WT: 65% vs Dyt1: 35%). Trihexyphenidyl required nicotinic receptors but not glutamate receptors to increase dopamine release. Dyt1 mice were more sensitive to the dopamine release decreasing effects of nicotinic acetylcholine receptor antagonism (IC50: WT = 29.46 nM, Dyt1 = 12.26 nM) and less sensitive to acetylcholinesterase inhibitors suggesting that nicotinic acetylcholine receptor neurotransmission is altered in Dyt1 mice, that nicotinic receptors indirectly mediate the differential effects of trihexyphenidyl in Dyt1 mice, and that nicotinic receptors may be suitable therapeutic targets for DYT1 dystonia.
Collapse
Affiliation(s)
- Anthony M Downs
- Department of Pharmacology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - Xueliang Fan
- Department of Pharmacology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - Christine Donsante
- Department of Pharmacology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA
| | - H A Jinnah
- Department of Neurology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA; Department of Human Genetics, Emory University School of Medicine, 101 Woodruff Circle, WMB 6300, Atlanta, GA 30322, USA; Department of Pediatrics, Emory University School of Medicine, 101 Woodruff Circle, WMB 6300, Atlanta, GA 30322, USA
| | - Ellen J Hess
- Department of Pharmacology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA; Department of Neurology, Emory University School of Medicine, 101 Woodruff Circle, WMB 6304, Atlanta, GA 30322, USA.
| |
Collapse
|
35
|
Fahn S, Sulzer D, Kang UJ, Bressman S. In memoriam: Robert E. Burke, MD, 1949–2018. Mov Disord 2019. [DOI: 10.1002/mds.27612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Stanley Fahn
- Division of Movement Disorders, Department of Neurology Columbia University Irving Medical Center New York New York USA
| | - David Sulzer
- Division of Movement Disorders, Department of Neurology Columbia University Irving Medical Center New York New York USA
- Departments of Psychiatry, Neurology and Pharmacology Columbia University Irving Medical Center New York New York USA
| | - Un Jung Kang
- Division of Movement Disorders, Department of Neurology Columbia University Irving Medical Center New York New York USA
| | - Susan Bressman
- Department of Neurology, Beth Israel Campus Mount Sinai Medical Center New York New York USA
| |
Collapse
|
36
|
Abudukeyoumu N, Hernandez-Flores T, Garcia-Munoz M, Arbuthnott GW. Cholinergic modulation of striatal microcircuits. Eur J Neurosci 2018; 49:604-622. [PMID: 29797362 PMCID: PMC6587740 DOI: 10.1111/ejn.13949] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/30/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022]
Abstract
The purpose of this review is to bridge the gap between earlier literature on striatal cholinergic interneurons and mechanisms of microcircuit interaction demonstrated with the use of newly available tools. It is well known that the main source of the high level of acetylcholine in the striatum, compared to other brain regions, is the cholinergic interneurons. These interneurons provide an extensive local innervation that suggests they may be a key modulator of striatal microcircuits. Supporting this idea requires the consideration of functional properties of these interneurons, their influence on medium spiny neurons, other interneurons, and interactions with other synaptic regulators. Here, we underline the effects of intrastriatal and extrastriatal afferents onto cholinergic interneurons and discuss the activation of pre‐ and postsynaptic muscarinic and nicotinic receptors that participate in the modulation of intrastriatal neuronal interactions. We further address recent findings about corelease of other transmitters in cholinergic interneurons and actions of these interneurons in striosome and matrix compartments. In addition, we summarize recent evidence on acetylcholine‐mediated striatal synaptic plasticity and propose roles for cholinergic interneurons in normal striatal physiology. A short examination of their role in neurological disorders such as Parkinson's, Huntington's, and Tourette's pathologies and dystonia is also included.
Collapse
Affiliation(s)
| | | | | | - Gordon W Arbuthnott
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
37
|
Long-Lasting Electrophysiological After-Effects of High-Frequency Stimulation in the Globus Pallidus: Human and Rodent Slice Studies. J Neurosci 2018; 38:10734-10746. [PMID: 30373767 DOI: 10.1523/jneurosci.0785-18.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/28/2018] [Accepted: 10/24/2018] [Indexed: 11/21/2022] Open
Abstract
Deep-brain stimulation (DBS) of the globus pallidus pars interna (GPi) is a highly effective therapy for movement disorders, yet its mechanism of action remains controversial. Inhibition of local neurons because of release of GABA from afferents to the GPi is a proposed mechanism in patients. Yet, high-frequency stimulation (HFS) produces prolonged membrane depolarization mediated by cholinergic neurotransmission in endopeduncular nucleus (EP, GPi equivalent in rodent) neurons. We applied HFS while recording neuronal firing from an adjacent electrode during microelectrode mapping of GPi in awake patients (both male and female) with Parkinson disease (PD) and dystonia. Aside from after-suppression and no change in neuronal firing, high-frequency microstimulation induced after-facilitation in 38% (26/69) of GPi neurons. In neurons displaying after-facilitation, 10 s HFS led to an immediate decrease of bursting in PD, but not dystonia patients. Moreover, the changes of bursting patterns in neurons with after-suppression or no change after HFS, were similar in both patient groups. To explore the mechanisms responsible, we applied HFS in EP brain slices from rats of either sex. As in humans, HFS in EP induced two subtypes of after-excitation: excitation or excitation with late inhibition. Pharmacological experiments determined that the excitation subtype, induced by lower charge density, was dependent on glutamatergic transmission. HFS with higher charge density induced excitation with late inhibition, which involved cholinergic modulation. Therefore HFS with different charge density may affect the local neurons through multiple synaptic mechanisms. The cholinergic system plays a role in mediating the after-facilitatory effects in GPi neurons, and because of their modulatory nature, may provide a basis for both the immediate and delayed effects of GPi-DBS. We propose a new model to explain the mechanisms of DBS in GPi.SIGNIFICANCE STATEMENT Deep-brain stimulation (DBS) in the globus pallidus pars interna (GPi) improves Parkinson disease (PD) and dystonia, yet its mechanisms in GPi remain controversial. Inhibition has been previously described and thought to indicate activation of GABAergic synaptic terminals, which dominate in GPi. Here we report that 10 s high-frequency microstimulation induced after-facilitation of neural firing in a substantial proportion of GPi neurons in humans. The neurons with after-facilitation, also immediately reduced their bursting activities after high-frequency stimulation in PD, but not dystonia patients. Based on these data and further animal experiments, a mechanistic hypothesis involving glutamatergic, GABAergic, and cholinergic synaptic transmission is proposed to explain both short- and longer-term therapeutic effects of DBS in GPi.
Collapse
|
38
|
Abstract
Dystonia is a neurological condition characterized by abnormal involuntary movements or postures owing to sustained or intermittent muscle contractions. Dystonia can be the manifesting neurological sign of many disorders, either in isolation (isolated dystonia) or with additional signs (combined dystonia). The main focus of this Primer is forms of isolated dystonia of idiopathic or genetic aetiology. These disorders differ in manifestations and severity but can affect all age groups and lead to substantial disability and impaired quality of life. The discovery of genes underlying the mendelian forms of isolated or combined dystonia has led to a better understanding of its pathophysiology. In some of the most common genetic dystonias, such as those caused by TOR1A, THAP1, GCH1 and KMT2B mutations, and idiopathic dystonia, these mechanisms include abnormalities in transcriptional regulation, striatal dopaminergic signalling and synaptic plasticity and a loss of inhibition at neuronal circuits. The diagnosis of dystonia is largely based on clinical signs, and the diagnosis and aetiological definition of this disorder remain a challenge. Effective symptomatic treatments with pharmacological therapy (anticholinergics), intramuscular botulinum toxin injection and deep brain stimulation are available; however, future research will hopefully lead to reliable biomarkers, better treatments and cure of this disorder.
Collapse
|
39
|
Pappas SS, Li J, LeWitt TM, Kim JK, Monani UR, Dauer WT. A cell autonomous torsinA requirement for cholinergic neuron survival and motor control. eLife 2018; 7:36691. [PMID: 30117805 PMCID: PMC6115190 DOI: 10.7554/elife.36691] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 08/16/2018] [Indexed: 12/14/2022] Open
Abstract
Cholinergic dysfunction is strongly implicated in dystonia pathophysiology. Previously (Pappas et al., 2015;4:e08352), we reported that Dlx5/6-Cre mediated forebrain deletion of the DYT1 dystonia protein torsinA (Dlx-CKO) causes abnormal twisting and selective degeneration of dorsal striatal cholinergic interneurons (ChI) (Pappas et al., 2015). A central question raised by that work is whether the ChI loss is cell autonomous or requires torsinA loss from neurons synaptically connected to ChIs. Here, we addressed this question by using ChAT-Cre mice to conditionally delete torsinA from cholinergic neurons ('ChAT-CKO'). ChAT-CKO mice phenocopy the Dlx-CKO phenotype of selective dorsal striatal ChI loss and identify an essential requirement for torsinA in brainstem and spinal cholinergic neurons. ChAT-CKO mice are tremulous, weak, and exhibit trunk twisting and postural abnormalities. These findings are the first to demonstrate a cell autonomous requirement for torsinA in specific populations of cholinergic neurons, strengthening the connection between torsinA, cholinergic dysfunction and dystonia pathophysiology.
Collapse
Affiliation(s)
- Samuel S Pappas
- Department of Neurology, University of Michigan, Ann Arbor, United States
| | - Jay Li
- Department of Neurology, University of Michigan, Ann Arbor, United States.,Cell and Molecular Biology Program, University of Michigan, Ann Arbor, United States
| | - Tessa M LeWitt
- Department of Neurology, University of Michigan, Ann Arbor, United States
| | - Jeong-Ki Kim
- Department of Cell Biology, Columbia University Medical Center, New York, United States.,Center for Motor Neuron Biology and Disease, Columbia University Medical Center, New York, United States.,Department of Pathology, Columbia University Medical Center, New York, United States
| | - Umrao R Monani
- Department of Cell Biology, Columbia University Medical Center, New York, United States.,Center for Motor Neuron Biology and Disease, Columbia University Medical Center, New York, United States.,Department of Pathology, Columbia University Medical Center, New York, United States
| | - William T Dauer
- Department of Neurology, University of Michigan, Ann Arbor, United States.,Cell and Molecular Biology Program, University of Michigan, Ann Arbor, United States.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, United States
| |
Collapse
|
40
|
Vijayakumar D, Jankovic J. Medical treatment of blepharospasm. EXPERT REVIEW OF OPHTHALMOLOGY 2018. [DOI: 10.1080/17469899.2018.1503535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Dhanya Vijayakumar
- The University of South Carolina School of Medicine Greenville, Neuroscience Associates/Department of Internal Medicine, Greenville Health System, Greenville, South Carolina, USA
| | - Joseph Jankovic
- Department of Neurology, Baylor College of Medicine, Baylor St. Luke’s Medical Center at the McNair Campus, Houston, Texas, USA
| |
Collapse
|
41
|
Tailor the pharmacological management of childhood dystonia to meet the needs of each child. DRUGS & THERAPY PERSPECTIVES 2018. [DOI: 10.1007/s40267-018-0495-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
42
|
Russ JB, Nallappan AM, Robichaux-Viehoever A. Management of Pediatric Movement Disorders: Present and Future. Semin Pediatr Neurol 2018; 25:136-151. [PMID: 29735111 DOI: 10.1016/j.spen.2018.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Management of movement disorders in children is an evolving field. This article outlines the major categories of treatment options for pediatric movement disorders and general guidelines for their use. We review the evidence for existing therapies, which continue to lack large-scale controlled trials to guide treatment decisions. The field continues to rely on extrapolations from adult studies and lower quality evidence such as case reports and case series to guide treatment guidelines and consensus statements. Developments in new pharmaceuticals for rare diseases have begun to provide hope for those cases in which a genetic diagnosis can be made. Advances in surgical therapies such as deep brain stimulation as well as new modes of treatment such as gene therapy, epigenetic modulation, and stem cell therapy hold promise for improving outcomes in both primary and secondary causes of movement disorders. There is a critical need for larger, multicenter, controlled clinical trials to fully evaluate treatments for pediatric movement disorders.
Collapse
Affiliation(s)
- Jeffrey B Russ
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Akila M Nallappan
- Undergraduate Program, Case Western Reserve University, Cleveland, OH
| | | |
Collapse
|
43
|
Putzel GG, Battistella G, Rumbach AF, Ozelius LJ, Sabuncu MR, Simonyan K. Polygenic Risk of Spasmodic Dysphonia is Associated With Vulnerable Sensorimotor Connectivity. Cereb Cortex 2018; 28:158-166. [PMID: 29117296 PMCID: PMC6059246 DOI: 10.1093/cercor/bhw363] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 10/28/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022] Open
Abstract
Spasmodic dysphonia (SD), or laryngeal dystonia, is an isolated task-specific dystonia of unknown causes and pathophysiology that selectively affects speech production. Using next-generation whole-exome sequencing in SD patients, we computed polygenic risk score from 1804 genetic markers based on a genome-wide association study in another form of similar task-specific focal dystonia, musician's dystonia. We further examined the associations between the polygenic risk score, resting-state functional connectivity abnormalities within the sensorimotor network, and SD clinical characteristics. We found that the polygenic risk of dystonia was significantly associated with decreased functional connectivity in the left premotor/primary sensorimotor and inferior parietal cortices in SD patients. Reduced connectivity of the inferior parietal cortex was correlated with the age of SD onset. The polygenic risk score contained a significant number of genetic variants lying near genes related to synaptic transmission and neural development. Our study identified a polygenic contribution to the overall genetic risk of dystonia in the cohort of SD patients. Associations between the polygenic risk and reduced functional connectivity of the sensorimotor and inferior parietal cortices likely represent an endophenotypic imaging marker of SD, while genes involved in synaptic transmission and neuron development may be linked to the molecular pathophysiology of this disorder.
Collapse
Affiliation(s)
- Gregory Garbès Putzel
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY10029, USA
| | - Giovanni Battistella
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY10029, USA
| | - Anna F Rumbach
- School of Health and Rehabilitation Sciences, Speech Pathology, University of Queensland, Brisbane, Queensland, QLD, 4072, Australia
| | - Laurie J Ozelius
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA02129, USA
| | - Mert R Sabuncu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA02129, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA02128, USA
| | - Kristina Simonyan
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY10029, USA
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY10029, USA
| |
Collapse
|
44
|
Masson R, Pagliano E, Baranello G. Efficacy of oral pharmacological treatments in dyskinetic cerebral palsy: a systematic review. Dev Med Child Neurol 2017; 59:1237-1248. [PMID: 28872668 DOI: 10.1111/dmcn.13532] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/15/2017] [Indexed: 12/19/2022]
Abstract
AIM To evaluate the actual evidence of efficacy of oral pharmacological treatments in the management of dyskinetic cerebral palsy (CP). METHOD A systematic review was performed according to the American Academy for Cerebral Palsy and Developmental Medicine (AACPDM) and Preferred Reporting Items for Systematic Reviews and Meta-Analyses methodology. Articles were searched for in PubMed/MEDLINE, Scopus, Web of Science, Cochrane Library, Database of Reviews of Effectiveness, OTSeeker, Physiotherapy Evidence Database, REHABDATA, and ClinicalTrials.gov. RESULTS Sixteen articles met the eligibility criteria. Eight studies on trihexyphenidyl and two on levodopa showed contradictory results. Low efficacy was reported for diazepam, dantrolene sodium, perphenazine, and etybenzatropine. Tetrabenazine, gabapentin and levetiracetam should be studied in more detail. The updated available evidence does not support any therapeutic algorithm for the management of dyskinetic CP. INTERPRETATION This lack of evidence is partially owing to the inconsistency of classifications of patients and of outcome measures used in the reviewed studies. Further randomized, double-blind, placebo-controlled pharmacological trials, optimized for different age groups, based on valid, reliable, and disease-specific rating scales are strongly needed. Outcome measures should be selected within the framework of the International Classification of Functioning, Disability and Health. WHAT THIS PAPER ADDS Evidence to prove (or disprove) the efficacy of oral drugs in dyskinetic cerebral palsy is low. The most investigated drugs, trihexyphenidyl and levodopa, show contradictory results. Tetrabenazine, levetiracetam, and gabapentin efficacy should be studied in more detail. Lack of evidence is partially due to the inconsistency of classifications and outcome measures used. Outcome measures should be selected within the framework of the International Classification of Functioning, Disability and Health in next clinical trials.
Collapse
Affiliation(s)
- Riccardo Masson
- Developmental Neurology Unit, "Carlo Besta" Neurological Institute Foundation, Milan, Italy
| | - Emanuela Pagliano
- Developmental Neurology Unit, "Carlo Besta" Neurological Institute Foundation, Milan, Italy
| | - Giovanni Baranello
- Developmental Neurology Unit, "Carlo Besta" Neurological Institute Foundation, Milan, Italy
| |
Collapse
|
45
|
Advances in pharmacotherapies for movement disorders in children: current limitations and future progress. Curr Opin Pediatr 2017; 29:652-664. [PMID: 29120894 DOI: 10.1097/mop.0000000000000555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW In childhood, movement disorders are generated by a very large number of disorders of the nervous system, and the very different developmental ages at which these occur make studies of pharmacotherapy efficacy extremely difficult. In most clinical practices, medication used in management is by trial and error, and limited by lack of efficacy and/or adverse drug reactions leading to drug intolerance. Nevertheless, symptom reduction using polypharmacy must be balanced against any accompanying comorbidities such as poor attention and concentration, constipation, ileus, urinary retention, blurred vision sedation and respiratory depression. RECENT FINDINGS A 'personalised medicine' approach may lead to specific management breakthroughs that are beneficial to a wider number of children. At present, neuromodulation with implantable devices offers greater proven efficacy for dystonia, myoclonus and dystonic-choreoathetosis, but enteral, intravenous and, more recently, transdermal medication strategies with clonidine patches and enteral gabapentin may provide important relief for both home management and critical care settings. SUMMARY The current review brings the clinician up-to-date with the latest, albeit limited, thinking on the pharmacological management of movement disorders in children by focussing on goal-directed outcome measures to improve clinical decision-making in an evidence-light clinical setting.
Collapse
|
46
|
Abstract
Dystonia is one of the most frequent movement disorders in childhood. It can impede normal motor development and cause significant motor disability. The diagnostic evaluation of childhood dystonia is challenging due to the phenotypic variability and heterogeneous etiologies. Evidence to guide the diagnostic evaluation and treatment is limited. Assessment is primarily directed by clinical history and distinctive examination findings. Neuroimaging is typically necessary to evaluate for acquired or complex inherited dystonias. A trial of levodopa can be both diagnostic and therapeutic in children with dopa-responsive dystonia. However, for the majority of children with early-onset dystonia, treatment is symptomatic with varying efficacy. There is a paucity of therapeutic trials for childhood dystonia and most treatment recommendations are consensus or expert opinion driven. This review summarizes the available evidence and guidelines on the diagnostic evaluation and pharmacological treatment of childhood-onset dystonia and provides practical frameworks to approach both issues based on best evidence.
Collapse
|
47
|
Burciu RG, Hess CW, Coombes SA, Ofori E, Shukla P, Chung JW, McFarland NR, Wagle Shukla A, Okun MS, Vaillancourt DE. Functional activity of the sensorimotor cortex and cerebellum relates to cervical dystonia symptoms. Hum Brain Mapp 2017; 38:4563-4573. [PMID: 28594097 PMCID: PMC5547035 DOI: 10.1002/hbm.23684] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 12/31/2022] Open
Abstract
Cervical dystonia (CD) is the most common type of focal dystonia, causing abnormal movements of the neck and head. In this study, we used noninvasive imaging to investigate the motor system of patients with CD and uncover the neural correlates of dystonic symptoms. Furthermore, we examined whether a commonly prescribed anticholinergic medication in CD has an effect on the dystonia-related brain abnormalities. Participants included 16 patients with CD and 16 healthy age-matched controls. We collected functional MRI scans during a force task previously shown to extensively engage the motor system, and diffusion and T1-weighted MRI scans from which we calculated free-water and brain tissue densities. The dystonia group was also scanned ca. 2 h after a 2-mg dose of trihexyphenidyl. Severity of dystonia was assessed pre- and post-drug using the Burke-Fahn-Marsden Dystonia Rating Scale. Motor-related activity in CD was altered relative to controls in the primary somatosensory cortex, cerebellum, dorsal premotor and posterior parietal cortices, and occipital cortex. Most importantly, a regression model showed that increased severity of symptoms was associated with decreased functional activity of the somatosensory cortex and increased activity of the cerebellum. Structural imaging measures did not differ between CD and controls. The single dose of trihexyphenidyl altered the fMRI signal in the somatosensory cortex but not in the cerebellum. Symptom severity was not significantly reduced post-treatment. Findings show widespread changes in functional brain activity in CD and most importantly that dystonic symptoms relate to disrupted activity in the somatosensory cortex and cerebellum. Hum Brain Mapp 38:4563-4573, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Roxana G. Burciu
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFlorida
| | - Christopher W. Hess
- Department of NeurologyUniversity of FloridaGainesvilleFlorida
- Center for Movement Disorders and Neurorestoration, University of FloridaGainesvilleFlorida
| | - Stephen A. Coombes
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFlorida
| | - Edward Ofori
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFlorida
| | - Priyank Shukla
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFlorida
| | - Jae Woo Chung
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFlorida
| | - Nikolaus R. McFarland
- Department of NeurologyUniversity of FloridaGainesvilleFlorida
- Center for Movement Disorders and Neurorestoration, University of FloridaGainesvilleFlorida
| | - Aparna Wagle Shukla
- Department of NeurologyUniversity of FloridaGainesvilleFlorida
- Center for Movement Disorders and Neurorestoration, University of FloridaGainesvilleFlorida
| | - Michael S. Okun
- Department of NeurologyUniversity of FloridaGainesvilleFlorida
- Center for Movement Disorders and Neurorestoration, University of FloridaGainesvilleFlorida
- Department of NeurosurgeryUniversity of FloridaGainesvilleFlorida
| | - David E. Vaillancourt
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFlorida
- Department of NeurologyUniversity of FloridaGainesvilleFlorida
- Department of Biomedical EngineeringUniversity of FloridaGainesvilleFlorida
| |
Collapse
|
48
|
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
|
49
|
Affiliation(s)
- Isabel Alfradique-Dunham
- Parkinson’s Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph Jankovic
- Parkinson’s Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
50
|
Fasano A, Llinas M, Munhoz RP, Hlasny E, Kucharczyk W, Lozano AM. MRI-guided focused ultrasound thalamotomy in non-ET tremor syndromes. Neurology 2017; 89:771-775. [PMID: 28747452 DOI: 10.1212/wnl.0000000000004268] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 05/30/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To report the 6-month single-blinded results of unilateral thalamotomy with MRI-guided focused ultrasound (MRgFUS) in patients with tremors other than essential tremor. METHODS Three patients with tremor due to Parkinson disease, 2 with dystonic tremor in the context of cervicobrachial dystonia and writer's cramp, and 1 with dystonia gene-associated tremor underwent MRgFUS targeting the ventro-intermedius nucleus (Vim) of the dominant hemisphere. The primary endpoint was the reduction of lateralized items of the Tremor Rating Scale of contralateral hemibody assessed by a blinded rater. RESULTS All patients achieved a statistically significant, immediate, and sustained improvement of the contralateral tremor score by 42.2%, 52.0%, 55.9%, and 52.9% at 1 week and 1, 3, and 6 months after the procedure, respectively. All patients experienced transient side effects and 2 patients experienced persistent side effects at the time of last evaluation: hemitongue numbness and hemiparesis with hemihypoesthesia. CONCLUSIONS Vim MRgFUS is a promising, incision-free, but nevertheless invasive technique to effectively treat tremors other than essential tremor. Future studies on larger samples and longer follow-up will further define its effectiveness and safety. CLINICALTRIALSGOV IDENTIFIER NCT02252380. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that for patients with tremor not caused by essential tremor, MRgFUS of the Vim improves the tremor of the contralateral hemibody at 6 months.
Collapse
Affiliation(s)
- Alfonso Fasano
- From the Morton and Gloria Shulman Movement Disorders Clinic and Edmond J. Safra Program in Parkinson's Disease (A.F., R.P.M.), Toronto Western Hospital-UHN, Division of Neurology, University of Toronto; Krembil Research Institute (A.F.); Division of Neurosurgery (M.L., A.M.L.), Department of Surgery, Krembil Neuroscience Centre, Toronto Western Hospital, University of Toronto; Joint Department of Medical Imaging (E.H., W.K.), University Health Network; and Department of Medical Imaging (W.K.), University of Toronto, Ontario, Canada.
| | - Maheleth Llinas
- From the Morton and Gloria Shulman Movement Disorders Clinic and Edmond J. Safra Program in Parkinson's Disease (A.F., R.P.M.), Toronto Western Hospital-UHN, Division of Neurology, University of Toronto; Krembil Research Institute (A.F.); Division of Neurosurgery (M.L., A.M.L.), Department of Surgery, Krembil Neuroscience Centre, Toronto Western Hospital, University of Toronto; Joint Department of Medical Imaging (E.H., W.K.), University Health Network; and Department of Medical Imaging (W.K.), University of Toronto, Ontario, Canada
| | - Renato P Munhoz
- From the Morton and Gloria Shulman Movement Disorders Clinic and Edmond J. Safra Program in Parkinson's Disease (A.F., R.P.M.), Toronto Western Hospital-UHN, Division of Neurology, University of Toronto; Krembil Research Institute (A.F.); Division of Neurosurgery (M.L., A.M.L.), Department of Surgery, Krembil Neuroscience Centre, Toronto Western Hospital, University of Toronto; Joint Department of Medical Imaging (E.H., W.K.), University Health Network; and Department of Medical Imaging (W.K.), University of Toronto, Ontario, Canada
| | - Eugen Hlasny
- From the Morton and Gloria Shulman Movement Disorders Clinic and Edmond J. Safra Program in Parkinson's Disease (A.F., R.P.M.), Toronto Western Hospital-UHN, Division of Neurology, University of Toronto; Krembil Research Institute (A.F.); Division of Neurosurgery (M.L., A.M.L.), Department of Surgery, Krembil Neuroscience Centre, Toronto Western Hospital, University of Toronto; Joint Department of Medical Imaging (E.H., W.K.), University Health Network; and Department of Medical Imaging (W.K.), University of Toronto, Ontario, Canada
| | - Walter Kucharczyk
- From the Morton and Gloria Shulman Movement Disorders Clinic and Edmond J. Safra Program in Parkinson's Disease (A.F., R.P.M.), Toronto Western Hospital-UHN, Division of Neurology, University of Toronto; Krembil Research Institute (A.F.); Division of Neurosurgery (M.L., A.M.L.), Department of Surgery, Krembil Neuroscience Centre, Toronto Western Hospital, University of Toronto; Joint Department of Medical Imaging (E.H., W.K.), University Health Network; and Department of Medical Imaging (W.K.), University of Toronto, Ontario, Canada
| | - Andres M Lozano
- From the Morton and Gloria Shulman Movement Disorders Clinic and Edmond J. Safra Program in Parkinson's Disease (A.F., R.P.M.), Toronto Western Hospital-UHN, Division of Neurology, University of Toronto; Krembil Research Institute (A.F.); Division of Neurosurgery (M.L., A.M.L.), Department of Surgery, Krembil Neuroscience Centre, Toronto Western Hospital, University of Toronto; Joint Department of Medical Imaging (E.H., W.K.), University Health Network; and Department of Medical Imaging (W.K.), University of Toronto, Ontario, Canada
| |
Collapse
|