1
|
Goto S. Functional pathology of neuroleptic-induced dystonia based on the striatal striosome-matrix dopamine system in humans. J Neurol Neurosurg Psychiatry 2024:jnnp-2024-334545. [PMID: 39631787 DOI: 10.1136/jnnp-2024-334545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
Neuroleptic-induced dystonia is a source of great concern in clinical practice because of its iatrogenic nature which can potentially lead to life-threatening conditions. Since all neuroleptics (antipsychotics) share the ability to block the dopamine D2-type receptors (D2Rs) that are highly enriched in the striatum, this drug-induced dystonia is thought to be caused by decreased striatal D2R activity. However, how associations of striatal D2R inactivation with dystonia are formed remains elusive.A growing body of evidence suggests that imbalanced activities between D1R-expressing medium spiny neurons and D2R-expressing medium spiny neurons (D1-MSNs and D2-MSNs) in the striatal striosome-matrix system underlie the pathophysiology of various basal ganglia disorders including dystonia. Given the specificity of the striatal dopamine D1 system in 'humans', this article highlights the striatal striosome hypothesis in causing 'repetitive' and 'stereotyped' motor symptoms which are key clinical features of dystonia. It is suggested that exposure to neuroleptics may reduce striosomal D1-MSN activity and thereby cause dystonia symptoms. This may occur through an increase in the striatal cholinergic activity and the collateral inhibitory action of D2-MSNs onto neighbouring D1-MSNs within the striosome subfields. The article proposes a functional pathology of the striosome-matrix dopamine system for neuroleptic-induced acute dystonia or neuroleptic-withdrawal dystonia. A rationale for the effectiveness of dopaminergic or cholinergic pharmacotherapy is also provided for treating dystonias. This narrative review covers various aspects of the relevant field and provides a detailed discussion of the mechanisms of neuroleptic-induced dystonia.
Collapse
Affiliation(s)
- Satoshi Goto
- Research Organization of Science and Technology, Ritsumeikan University, Kyoto, Japan
| |
Collapse
|
2
|
Calakos N, Zech M. Emerging Molecular-Genetic Families in Dystonia: Endosome-Autophagosome-Lysosome and Integrated Stress Response Pathways. Mov Disord 2024. [PMID: 39467044 DOI: 10.1002/mds.30037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
Advances in genetic technologies and disease modeling have greatly accelerated the pace of introducing and validating molecular-genetic contributors to disease. In dystonia, there is a growing convergence across multiple distinct forms of the disease onto core biological processes. Here, we discuss two of these, the endosome-autophagosome-lysosome pathway and the integrated stress response, to highlight recent advances in the field. Using these two pathomechanisms as examples, we further discuss the opportunities that molecular-genetic grouping of dystonias present to transform dystonia care. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Nicole Calakos
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Michael Zech
- Institute of Human Genetics, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Institute of Neurogenomics, Helmholtz Munich, Neuherberg, Germany
- Institute for Advanced Study, Technical University of Munich, Garching, Germany
| |
Collapse
|
3
|
Gemperli K, Folorunso F, Norin B, Joshua R, Rykowski R, Hill C, Galindo R, Aravamuthan BR. Preterm birth is associated with dystonic features and reduced cortical parvalbumin immunoreactivity in mice. Pediatr Res 2024:10.1038/s41390-024-03603-8. [PMID: 39433959 DOI: 10.1038/s41390-024-03603-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/19/2024] [Accepted: 09/15/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND Preterm birth is a common cause of dystonia. Though dystonia is often associated with striatal dysfunction after neonatal brain injury, cortical dysfunction may best predict dystonia following preterm birth. Furthermore, abnormal sensorimotor cortex inhibition is associated with genetic and idiopathic dystonias. To investigate cortical dysfunction and dystonia following preterm birth, we developed a new model of preterm birth in mice. METHODS We induced preterm birth in C57BL/6J mice at embryonic day 18.3, ~24 h early. Leg adduction variability and amplitude, metrics we have shown distinguish between dystonia from spasticity during gait in people with CP, were quantified from gait videos of mice. Parvalbumin-positive interneurons, the largest population of cortical inhibitory interneurons, were quantified in the sensorimotor cortex and striatum. RESULTS Mice born preterm demonstrate increased leg adduction amplitude and variability during gait, suggestive of clinically observed dystonic gait features. Mice born preterm also demonstrate fewer parvalbumin-positive interneurons and reduced parvalbumin immunoreactivity in the sensorimotor cortex, but not the striatum, suggesting dysfunction of cortical inhibition. CONCLUSIONS These data may suggest an association between cortical dysfunction and dystonic gait features following preterm birth. We propose that our novel mouse model of preterm birth can be used to study this association. IMPACT Mouse models of true preterm birth are valuable for studying clinical complications of prematurity. Mice born preterm demonstrate increased leg adduction amplitude and variability during gait, suggestive of clinically observed dystonic gait features. Mice born preterm demonstrate fewer parvalbumin-positive interneurons and reduced parvalbumin immunoreactivity in the sensorimotor cortex, suggesting dysfunction of cortical inhibition. Mice born preterm do not demonstrate changes in parvalbumin immunoreactivity in the striatum. Cortical dysfunction may be associated with dystonic gait features following preterm birth.
Collapse
Affiliation(s)
- Kat Gemperli
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Femi Folorunso
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Benjamin Norin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rebecca Joshua
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel Rykowski
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Clayton Hill
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rafael Galindo
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bhooma R Aravamuthan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
4
|
Scarduzio M, Eskow Jaunarajs KL, Standaert DG. Striatal cholinergic transmission in an inducible transgenic mouse model of paroxysmal non-kinesiogenic dyskinesia. Neurobiol Dis 2024; 201:106685. [PMID: 39343248 DOI: 10.1016/j.nbd.2024.106685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024] Open
Abstract
Altered interaction between striatonigral dopaminergic (DA) inputs and local acetylcholine (ACh) in striatum has long been hypothesized to play a central role in the pathophysiology of dystonia and dyskinesia. Indeed, previous research using several genetic mouse models of human isolated dystonia identified a shared endophenotype with paradoxical excitation of striatal cholinergic interneuron (ChIs) activity in response to activation of dopamine D2 receptors (D2R). These mouse models lack a dystonic motor phenotype, which leaves a critical gap in comprehending the role of DA and ACh transmission in the manifestations of dystonia. To tackle this question, we used a combination of ex vivo slice physiology and in vivo monitoring of striatal ACh dynamics in the inducible, phenotypically penetrant, transgenic mouse model of paroxysmal non-kinesiogenic dyskinesia (PNKD), an animal with both dystonic and dyskinetic features. We found that, similarly to genetic models of isolated dystonia, the PNKD mouse displays D2R-induced paradoxical excitation of ChI firing in ex vivo striatal brain slices. In vivo, caffeine triggers dystonic symptoms while reversing the D2R-mediated excitation of ChIs and desynchronizing ACh release in PNKD mice. In WT littermate controls, caffeine stimulates spontaneous locomotion through a similar but reversed mechanism involving an excitatory switch of the D2R control of ChI activity, associated with enhanced synchronization of ACh release. These observations suggest that the "paradoxical excitation" of cholinergic interneurons described in isolated dystonia models could represent a compensatory or protective mechanism that prevents manifestation of movement abnormalities and that phenotypic dystonia is possible only when this is absent. These findings also suggest that D2Rs may play an important role in synchronizing the ChI network leading to rhythmic ACh release during heightened movement states. Dysfunction of this interaction and corresponding desynchrony of ACh release may contribute to aberrant movements.
Collapse
Affiliation(s)
- Mariangela Scarduzio
- Center for Neurodegeneration and Experimental Therapeutics, Birmingham, AL, USA; Department of Neurology, UAB, Birmingham, AL, USA.
| | - Karen L Eskow Jaunarajs
- Center for Neurodegeneration and Experimental Therapeutics, Birmingham, AL, USA; Department of Neurology, UAB, Birmingham, AL, USA
| | - David G Standaert
- Center for Neurodegeneration and Experimental Therapeutics, Birmingham, AL, USA; Department of Neurology, UAB, Birmingham, AL, USA
| |
Collapse
|
5
|
Duhne M, Mohebi A, Kim K, Pelattini L, Berke JD. A mismatch between striatal cholinergic pauses and dopaminergic reward prediction errors. Proc Natl Acad Sci U S A 2024; 121:e2410828121. [PMID: 39365823 PMCID: PMC11474027 DOI: 10.1073/pnas.2410828121] [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: 05/31/2024] [Accepted: 08/23/2024] [Indexed: 10/06/2024] Open
Abstract
Striatal acetylcholine and dopamine critically regulate movement, motivation, and reward-related learning. Pauses in cholinergic interneuron (CIN) firing are thought to coincide with dopamine pulses encoding reward prediction errors (RPE) to jointly enable synaptic plasticity. Here, we examine the firing of identified CINs during reward-guided decision-making in freely moving rats and compare this firing to dopamine release. Relationships between CINs, dopamine, and behavior varied strongly by subregion. In the dorsal-lateral striatum, a Go! cue evoked burst-pause CIN spiking, followed by a brief dopamine pulse that was unrelated to RPE. In the dorsal-medial striatum, this cue evoked only a CIN pause, that was curtailed by a movement-selective rebound in firing. Finally, in the ventral striatum, a reward cue evoked RPE-coding increases in both dopamine and CIN firing, without a consistent pause. Our results demonstrate a spatial and temporal dissociation between CIN pauses and dopamine RPE signals and will inform future models of striatal information processing under both normal and pathological conditions.
Collapse
Affiliation(s)
- Mariana Duhne
- Department of Neurology, University of California, San Francisco, CA94158
| | - Ali Mohebi
- Department of Neurology, University of California, San Francisco, CA94158
| | - Kyoungjun Kim
- Department of Neurology, University of California, San Francisco, CA94158
| | - Lilian Pelattini
- Department of Neurology, University of California, San Francisco, CA94158
| | - Joshua D. Berke
- Department of Neurology, University of California, San Francisco, CA94158
- Department of Psychiatry and Behavioral Science, University of California, San Francisco, CA94107
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA94158
- Weill Institute for Neurosciences, University of California, San Francisco, CA94158
| |
Collapse
|
6
|
Chambers NE, Hall D, Barsoum S, Miller E, Curry T, Kaplan M, Garan S, Gallardo I, Staab R, Nabert D, Hutchinson K, Millett M, Moehle MS. Conditional Knockout of Striatal Gnal Produces Dystonia-like Motor Phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609754. [PMID: 39253490 PMCID: PMC11383043 DOI: 10.1101/2024.08.26.609754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Loss-of-function mutations in GNAL have been linked to an adult-onset, isolated dystonia that is largely indistinguishable from idiopathic dystonia. GNAL encodes Gα olf , a heterotrimeric G-protein α subunit with a defined molecular function to increase the production of the second messenger cAMP. Gα olf is abundant in the striatum, and is the only stimulatory G-protein in many cell types of the striatum. Due to the defined molecular signaling pathway and expression pattern of Gα olf , the clear genetic link to dystonia makes GNAL an exciting target to understand the pathological mechanisms of not only this genetic dystonia, but also the larger idiopathic disease. To better understand GNAL -linked dystonia, we generated a novel genetic mouse model that allows us to conditionally knock out Gnal in a site and time-specific manner. In the current study we used genetic or AAV based approaches to express Cre to knockout striatal Gnal in our novel Gnal fl/fl model. We then performed motor behavioral testing and ex vivo whole-cell patch clamp electrophysiology of striatal spiny projection neurons to interrogate how loss of Gnal leads to dystonia. Mice with conditional striatal knockout of Gnal show hindlimb clasping, other dystonia-like postures, less motor coordination, slowness, and torticollis as compared to age-matched controls. Furthermore, striatal spiny projection neurons show increased excitability in Gnal knockout animals. These exciting data are the first to report uninduced, overt dystonia in a mouse model of GNAL- linked dystonia, and directly correlate these with changes in spiny projection neuron electrophysiological properties. Our results show that adult loss of Gnal in the striatum leads to the development of dystonia, through homeostatic, paradoxical increases in spiny projection neuron excitability, and suggest that therapeutic strategies aimed at decreasing this hyperexcitable phenotype may provide symptomatic relief for patients with disease. One Sentence Summary: When Gnal is knocked out in the striatum of mice we observe overt behavioral symptoms and hyperexcitability in striatal spiny projection neurons.
Collapse
|
7
|
Lockshin ER, Calakos N. The integrated stress response in brain diseases: A double-edged sword for proteostasis and synapses. Curr Opin Neurobiol 2024; 87:102886. [PMID: 38901329 PMCID: PMC11646490 DOI: 10.1016/j.conb.2024.102886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024]
Abstract
The integrated stress response (ISR) is a highly conserved biochemical pathway that regulates protein synthesis. The ISR is activated in response to diverse stressors to restore cellular homeostasis. As such, the ISR is implicated in a wide range of diseases, including brain disorders. However, in the brain, the ISR also has potent influence on processes beyond proteostasis, namely synaptic plasticity, learning and memory. Thus, in the setting of brain diseases, ISR activity may have dual effects on proteostasis and synaptic function. In this review, we consider the ISR's contribution to brain disorders through the lens of its potential effects on synaptic plasticity. From these examples, we illustrate that at times ISR activity may be a "double-edged sword". We also highlight its potential as a therapeutic target to improve circuit function in brain diseases independent of its role in disease pathogenesis.
Collapse
Affiliation(s)
- Elana R Lockshin
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Nicole Calakos
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
8
|
Calakos N, Caffall ZF. The integrated stress response pathway and neuromodulator signaling in the brain: lessons learned from dystonia. J Clin Invest 2024; 134:e177833. [PMID: 38557486 PMCID: PMC10977992 DOI: 10.1172/jci177833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
The integrated stress response (ISR) is a highly conserved biochemical pathway involved in maintaining proteostasis and cell health in the face of diverse stressors. In this Review, we discuss a relatively noncanonical role for the ISR in neuromodulatory neurons and its implications for synaptic plasticity, learning, and memory. Beyond its roles in stress response, the ISR has been extensively studied in the brain, where it potently influences learning and memory, and in the process of synaptic plasticity, which is a substrate for adaptive behavior. Recent findings demonstrate that some neuromodulatory neuron types engage the ISR in an "always-on" mode, rather than the more canonical "on-demand" response to transient perturbations. Atypical demand for the ISR in neuromodulatory neurons introduces an additional mechanism to consider when investigating ISR effects on synaptic plasticity, learning, and memory. This basic science discovery emerged from a consideration of how the ISR might be contributing to human disease. To highlight how, in scientific discovery, the route from starting point to outcomes can often be circuitous and full of surprise, we begin by describing our group's initial introduction to the ISR, which arose from a desire to understand causes for a rare movement disorder, dystonia. Ultimately, the unexpected connection led to a deeper understanding of its fundamental role in the biology of neuromodulatory neurons, learning, and memory.
Collapse
Affiliation(s)
- Nicole Calakos
- Department of Neurology
- Department of Neurobiology, and
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | | |
Collapse
|
9
|
Gemperli K, Folorunso F, Norin B, Joshua R, Hill C, Rykowski R, Galindo R, Aravamuthan BR. Mice born preterm develop gait dystonia and reduced cortical parvalbumin immunoreactivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578353. [PMID: 38352408 PMCID: PMC10862908 DOI: 10.1101/2024.02.01.578353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Preterm birth leading to cerebral palsy (CP) is the most common cause of childhood dystonia, a movement disorder that is debilitating and often treatment refractory. Dystonia has been typically associated with dysfunction of striatal cholinergic interneurons, but clinical imaging data suggests that cortical injury may best predict dystonia following preterm birth. Furthermore, abnormal sensorimotor cortex inhibition has been found in many studies of non-CP dystonias. To assess the potential for a cortical etiology of dystonia following preterm birth, we developed a new model of preterm birth in mice. Noting that term delivery in mice on a C57BL/6J background is embryonic day 19.1 (E19.1), we induced preterm birth at the limits of pup viability at embryonic day (E) 18.3, equivalent to human 22 weeks gestation. Mice born preterm demonstrate display clinically validated metrics of dystonia during gait (leg adduction amplitude and variability) and also demonstrate reduced parvalbumin immunoreactivity in the sensorimotor cortex, suggesting dysfunction of cortical parvalbumin-positive inhibitory interneurons. Notably, reduced parvalbumin immunoreactivity or changes in parvalbumin-positive neuronal number were not observed in the striatum. These data support the association between cortical dysfunction and dystonia following preterm birth. We propose that our mouse model of preterm birth can be used to study this association and potentially also study other sequelae of extreme prematurity.
Collapse
Affiliation(s)
- Kat Gemperli
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Femi Folorunso
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Benjamin Norin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Rebecca Joshua
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Clayton Hill
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Rachel Rykowski
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | | | | |
Collapse
|
10
|
Gelineau-Morel R, Smyser C, Leeder JS. Identifying Effective Treatments for Dystonia in Patients With Cerebral Palsy: A Precision Therapeutics Approach. Neurology 2023; 101:752-759. [PMID: 37463749 PMCID: PMC10624496 DOI: 10.1212/wnl.0000000000207593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/12/2023] [Indexed: 07/20/2023] Open
Abstract
Recent focus on improving the recognition of dystonia in cerebral palsy (DCP) has highlighted the need for more effective treatments. Evidence supports improved functional outcomes with early interventions for patients with cerebral palsy, but it is not known which interventions are most effective for DCP. Current pharmacologic recommendations for DCP are based largely on anecdotal evidence, with medications demonstrating minimal to moderate improvements in dystonia and variable efficacy between patients. Patients, families, and clinicians have identified the need for new and improved treatments in DCP, naming this as the top research theme in a recent Neurology® publication. Precision therapeutics focuses on providing early effective interventions that are individualized to every patient and can guide research priorities to improve treatments for DCP. This commentary outlines current obstacles to improving treatment of DCP and addresses how precision therapeutics can address each of these obstacles through 4 key components: (1) identification of predictive biomarkers to select patients likely to develop DCP in the future and for whom early intervention may be appropriate to delay or prevent full manifestation of dystonia, (2) stratification of patients with DCP into subgroups according to shared features (clinical, functional, biochemical, etc) to provide a targeted intervention based on those shared features, (3) administration of an individualized dose of an effective intervention to ensure adequate concentrations of the therapeutic entity at the site of action, and (4) monitoring of objective biomarkers of response to intervention. With implementation of each of these components of precision therapeutics, new and more effective treatments for every person with DCP can be realized.
Collapse
Affiliation(s)
- Rose Gelineau-Morel
- From the Division of Neurology (R.G.-M.), Children's Mercy Kansas City; School of Medicine (R.G.-M., J.S.L.), University of Missouri-Kansas City; Department of Pediatrics (R.G.-M., J.S.L.), University of Kansas Medical Center, Kansas City; Department of Pediatrics (C.S.), Department of Neurology (C.S.), and Mallinckrodt Institute of Radiology (C.S.), Washington University in St. Louis; and Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation (J.S.L.), Children's Mercy Kansas City, MO.
| | - Christopher Smyser
- From the Division of Neurology (R.G.-M.), Children's Mercy Kansas City; School of Medicine (R.G.-M., J.S.L.), University of Missouri-Kansas City; Department of Pediatrics (R.G.-M., J.S.L.), University of Kansas Medical Center, Kansas City; Department of Pediatrics (C.S.), Department of Neurology (C.S.), and Mallinckrodt Institute of Radiology (C.S.), Washington University in St. Louis; and Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation (J.S.L.), Children's Mercy Kansas City, MO
| | - J Steven Leeder
- From the Division of Neurology (R.G.-M.), Children's Mercy Kansas City; School of Medicine (R.G.-M., J.S.L.), University of Missouri-Kansas City; Department of Pediatrics (R.G.-M., J.S.L.), University of Kansas Medical Center, Kansas City; Department of Pediatrics (C.S.), Department of Neurology (C.S.), and Mallinckrodt Institute of Radiology (C.S.), Washington University in St. Louis; and Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation (J.S.L.), Children's Mercy Kansas City, MO
| |
Collapse
|
11
|
Gemperli K, Lu X, Chintalapati K, Rust A, Bajpai R, Suh N, Blackburn J, Gelineau-Morel R, Kruer MC, Mingbundersuk D, O'Malley J, Tochen L, Waugh J, Wu S, Feyma T, Perlmutter J, Mennerick S, McCall J, Aravamuthan BR. Chronic striatal cholinergic interneuron excitation induces clinically-relevant dystonic behavior in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.19.549778. [PMID: 37503287 PMCID: PMC10370117 DOI: 10.1101/2023.07.19.549778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Dystonia is common, debilitating, often medically refractory, and difficult to diagnose. The gold standard for both clinical and mouse model dystonia evaluation is subjective assessment, ideally by expert consensus. However, this subjectivity makes translational quantification of clinically-relevant dystonia metrics across species nearly impossible. Many mouse models of genetic dystonias display abnormal striatal cholinergic interneuron excitation, but few display subjectively dystonic features. Therefore, whether striatal cholinergic interneuron pathology causes dystonia remains unknown. To address these critical limitations, we first demonstrate that objectively quantifiable leg adduction variability correlates with leg dystonia severity in people. We then show that chemogenetic excitation of striatal cholinergic interneurons in mice causes comparable leg adduction variability in mice. This clinically-relevant dystonic behavior in mice does not occur with acute excitation, but rather develops after 14 days of ongoing striatal cholinergic interneuron excitation. This requirement for prolonged excitation recapitulates the clinically observed phenomena of a delay between an inciting brain injury and subsequent dystonia manifestation and demonstrates a causative link between chronic striatal cholinergic interneuron excitation and clinically-relevant dystonic behavior in mice. Therefore, these results support targeting striatal ChIs for dystonia drug development and suggests early treatment in the window following injury but prior to dystonia onset. One Sentence Summary Chronic excitation of dorsal striatal cholinergic interneuron causes clinically-relevant dystonic phenotypes in mice.
Collapse
|
12
|
Rey Hipolito AG, van der Heijden ME, Sillitoe RV. Physiology of Dystonia: Animal Studies. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 169:163-215. [PMID: 37482392 DOI: 10.1016/bs.irn.2023.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Dystonia is currently ranked as the third most prevalent motor disorder. It is typically characterized by involuntary muscle over- or co-contractions that can cause painful abnormal postures and jerky movements. Dystonia is a heterogenous disorder-across patients, dystonic symptoms vary in their severity, body distribution, temporal pattern, onset, and progression. There are also a growing number of genes that are associated with hereditary dystonia. In addition, multiple brain regions are associated with dystonic symptoms in both genetic and sporadic forms of the disease. The heterogeneity of dystonia has made it difficult to fully understand its underlying pathophysiology. However, the use of animal models has been used to uncover the complex circuit mechanisms that lead to dystonic behaviors. Here, we summarize findings from animal models harboring mutations in dystonia-associated genes and phenotypic animal models with overt dystonic motor signs resulting from spontaneous mutations, neural circuit perturbations, or pharmacological manipulations. Taken together, an emerging picture depicts dystonia as a result of brain-wide network dysfunction driven by basal ganglia and cerebellar dysfunction. In the basal ganglia, changes in dopaminergic, serotonergic, noradrenergic, and cholinergic signaling are found across different animal models. In the cerebellum, abnormal burst firing activity is observed in multiple dystonia models. We are now beginning to unveil the extent to which these structures mechanistically interact with each other. Such mechanisms inspire the use of pre-clinical animal models that will be used to design new therapies including drug treatments and brain stimulation.
Collapse
Affiliation(s)
- Alejandro G Rey Hipolito
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, United States
| | - Meike E van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, United States
| | - Roy V Sillitoe
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States; Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States; Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, United States; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, United States.
| |
Collapse
|
13
|
Chambers NE, Millett M, Moehle MS. The muscarinic M4 acetylcholine receptor exacerbates symptoms of movement disorders. Biochem Soc Trans 2023; 51:691-702. [PMID: 37013974 PMCID: PMC10212540 DOI: 10.1042/bst20220525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/31/2022] [Accepted: 03/14/2023] [Indexed: 04/05/2023]
Abstract
Barbeau's seesaw hypothesis of dopamine-acetylcholine balance has predominated movement disorders literature for years. Both the simplicity of the explanation and the matching efficacy of anticholinergic treatment in movement disorders seem to support this hypothesis. However, evidence from translational and clinical studies in movement disorders indicates that many features of this simple balance are lost, broken, or absent from movement disorders models or in imaging studies of patients with these disorders. This review reappraises the dopamine-acetylcholine balance hypothesis in light of recent evidence and describes how the Gαi/o coupled muscarinic M4 receptor acts in opposition to dopamine signaling in the basal ganglia. We highlight how M4 signaling can ameliorate or exacerbate movement disorders symptoms and physiological correlates of these symptoms in specific disease states. Furthermore, we propose future directions for investigation of this mechanisms to fully understand the potential efficacy of M4 targeting therapeutics in movement disorders. Overall, initial evidence suggest that M4 is a promising pharmaceutical target to ameliorate motor symptoms of hypo- and hyper-dopaminergic disorders.
Collapse
Affiliation(s)
- Nicole E. Chambers
- Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Michael Millett
- Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| | - Mark S. Moehle
- Department of Pharmacology and Therapeutics and Center for Translational Research in Neurodegeneration, University of Florida College of Medicine, Gainesville, FL 32610, U.S.A
| |
Collapse
|
14
|
Berryman D, Barrett J, Liu C, Maugee C, Waldbaum J, Yi D, Xing H, Yokoi F, Saxena S, Li Y. Motor deficit and lack of overt dystonia in Dlx conditional Dyt1 knockout mice. Behav Brain Res 2023; 439:114221. [PMID: 36417958 PMCID: PMC10364669 DOI: 10.1016/j.bbr.2022.114221] [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: 08/24/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
DYT1 or DYT-TOR1A dystonia is early-onset generalized dystonia caused by a trinucleotide deletion of GAG in the TOR1A or DYT1 gene leads to the loss of a glutamic acid residue in the resulting torsinA protein. A mouse model with overt dystonia is of unique importance to better understand the DYT1 pathophysiology and evaluate preclinical drug efficacy. DYT1 dystonia is likely a network disorder involving multiple brain regions, particularly the basal ganglia. Tor1a conditional knockout in the striatum or cerebral cortex leads to motor deficits, suggesting the importance of corticostriatal connection in the pathogenesis of dystonia. Indeed, corticostriatal long-term depression impairment has been demonstrated in multiple targeted DYT1 mouse models. Pappas and colleagues developed a conditional knockout line (Dlx-CKO) that inactivated Tor1a in the forebrain and surprisingly displayed overt dystonia. We set out to validate whether conditional knockout affecting both cortex and striatum would lead to overt dystonia and whether machine learning-based video behavioral analysis could be used to facilitate high throughput preclinical drug screening. We generated Dlx-CKO mice and found no overt dystonia or motor deficits at 4 months. At 8 months, retesting revealed motor deficits in rotarod, beam walking, grip strength, and hyperactivity in the open field; however, no overt dystonia was visually discernible or through the machine learning-based video analysis. Consistent with other targeted DYT1 mouse models, we observed age-dependent deficits in the beam walking test, which is likely a better motor behavioral test for preclinical drug testing but more labor-intensive when overt dystonia is absent.
Collapse
Affiliation(s)
- David Berryman
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA; Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Jake Barrett
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Canna Liu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Christian Maugee
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA; Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Julien Waldbaum
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Daiyao Yi
- Herbert Wertheim College of Engineering, Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, USA
| | - Hong Xing
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fumiaki Yokoi
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Shreya Saxena
- Herbert Wertheim College of Engineering, Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, USA
| | - Yuqing Li
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA; Genetics Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
15
|
Li ZY, Tian WT, Huang XJ, Cao L. The Pathogenesis of Paroxysmal Kinesigenic Dyskinesia: Current Concepts. Mov Disord 2023; 38:537-544. [PMID: 36718795 DOI: 10.1002/mds.29326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 02/01/2023] Open
Abstract
Paroxysmal kinesigenic dyskinesia (PKD) is a movement disorder characterized by recurrent and transient episodes of involuntary movements, including dystonia, chorea, ballism, or a combination of these, which are typically triggered by sudden voluntary movement. Disturbance of the basal ganglia-thalamo-cortical circuit has long been considered the cause of involuntary movements. Impairment of the gating function of the basal ganglia can cause an aberrant output toward the thalamus, which in turn leads to excessive activation of the cerebral cortex. Structural and functional abnormalities in the basal ganglia, thalamus, and cortex and abnormal connections between these brain regions have been found in patients with PKD. Recent studies have highlighted the role of the cerebellum in PKD. Insufficient suppression from the cerebellar cortex to the deep cerebellar nuclei could lead to overexcitation of the thalamocortical pathway. Therefore, this literature review aims to provide a comprehensive overview of the current research progress to explore the neural circuits and pathogenesis of PKD and promote further understanding and outlook on the pathophysiological mechanism of movement disorders. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Zi-Yi Li
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wo-Tu Tian
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Jun Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Cao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
El Atiallah I, Bonsi P, Tassone A, Martella G, Biella G, Castagno AN, Pisani A, Ponterio G. Synaptic Dysfunction in Dystonia: Update From Experimental Models. Curr Neuropharmacol 2023; 21:2310-2322. [PMID: 37464831 PMCID: PMC10556390 DOI: 10.2174/1570159x21666230718100156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 07/20/2023] Open
Abstract
Dystonia, the third most common movement disorder, refers to a heterogeneous group of neurological diseases characterized by involuntary, sustained or intermittent muscle contractions resulting in repetitive twisting movements and abnormal postures. In the last few years, several studies on animal models helped expand our knowledge of the molecular mechanisms underlying dystonia. These findings have reinforced the notion that the synaptic alterations found mainly in the basal ganglia and cerebellum, including the abnormal neurotransmitters signalling, receptor trafficking and synaptic plasticity, are a common hallmark of different forms of dystonia. In this review, we focus on the major contribution provided by rodent models of DYT-TOR1A, DYT-THAP1, DYT-GNAL, DYT/ PARK-GCH1, DYT/PARK-TH and DYT-SGCE dystonia, which reveal that an abnormal motor network and synaptic dysfunction represent key elements in the pathophysiology of dystonia.
Collapse
Affiliation(s)
- Ilham El Atiallah
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Gerardo Biella
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Antonio N. Castagno
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Fondazione Mondino, Pavia, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Fondazione Mondino, Pavia, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
17
|
Cheng F, Zheng W, Barbuti PA, Bonsi P, Liu C, Casadei N, Ponterio G, Meringolo M, Admard J, Dording CM, Yu-Taeger L, Nguyen HP, Grundmann-Hauser K, Ott T, Houlden H, Pisani A, Krüger R, Riess O. DYT6 mutated THAP1 is a cell type dependent regulator of the SP1 family. Brain 2022; 145:3968-3984. [PMID: 35015830 DOI: 10.1093/brain/awac001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 12/01/2021] [Accepted: 12/13/2021] [Indexed: 11/12/2022] Open
Abstract
DYT6 dystonia is caused by mutations in the transcription factor THAP1. THAP1 knock-out or knock-in mouse models revealed complex gene expression changes, which are potentially responsible for the pathogenesis of DYT6 dystonia. However, how THAP1 mutations lead to these gene expression alterations and whether the gene expression changes are also reflected in the brain of THAP1 patients are still unclear. In this study we used epigenetic and transcriptomic approaches combined with multiple model systems [THAP1 patients' frontal cortex, THAP1 patients' induced pluripotent stem cell (iPSC)-derived midbrain dopaminergic neurons, THAP1 heterozygous knock-out rat model, and THAP1 heterozygous knock-out SH-SY5Y cell lines] to uncover a novel function of THAP1 and the potential pathogenesis of DYT6 dystonia. We observed that THAP1 targeted only a minority of differentially expressed genes caused by its mutation. THAP1 mutations lead to dysregulation of genes mainly through regulation of SP1 family members, SP1 and SP4, in a cell type dependent manner. Comparing global differentially expressed genes detected in THAP1 patients' iPSC-derived midbrain dopaminergic neurons and THAP1 heterozygous knock-out rat striatum, we observed many common dysregulated genes and 61 of them were involved in dystonic syndrome-related pathways, like synaptic transmission, nervous system development, and locomotor behaviour. Further behavioural and electrophysiological studies confirmed the involvement of these pathways in THAP1 knock-out rats. Taken together, our study characterized the function of THAP1 and contributes to the understanding of the pathogenesis of primary dystonia in humans and rats. As SP1 family members were dysregulated in some neurodegenerative diseases, our data may link THAP1 dystonia to multiple neurological diseases and may thus provide common treatment targets.
Collapse
Affiliation(s)
- Fubo Cheng
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Wenxu Zheng
- Institute for Ophthalmic Research Centre for Ophthalmology, University of Tuebingen, Tuebingen, Germany
| | - Peter Antony Barbuti
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Chang Liu
- Institute of Biology, University of Hohenheim, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
- NGS Competence Center Tuebingen, Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Jakob Admard
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
- NGS Competence Center Tuebingen, Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Claire Marie Dording
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Libo Yu-Taeger
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
- Department of Human Genetics, Faculty of Medicine, Ruhr University Bochum, Bochum, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Faculty of Medicine, Ruhr University Bochum, Bochum, Germany
| | | | - Thomas Ott
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS C. Mondino Foundation, Pavia, Italy
| | - Rejko Krüger
- Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
- Parkinson Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
- NGS Competence Center Tuebingen, Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
18
|
Gilbert LA, Fehlings DL, Gross P, Kruer MC, Kwan W, Mink JW, Shusterman M, Aravamuthan BR. Top 10 Research Themes for Dystonia in Cerebral Palsy: A Community-Driven Research Agenda. Neurology 2022; 99:237-245. [PMID: 35715199 PMCID: PMC9442618 DOI: 10.1212/wnl.0000000000200911] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 05/16/2022] [Indexed: 11/15/2022] Open
Abstract
Dystonia in cerebral palsy (DCP) is a common, debilitating, but understudied condition. The CP community (people with CP and caregivers) is uniquely equipped to help determine the research questions that best address their needs. We developed a community-driven DCP research agenda using the well-established James Lind Alliance methodology. CP community members, researchers, and clinicians were recruited through multiple advocacy, research, and professional organizations. To ensure shared baseline knowledge, participants watched webinars outlining our current knowledge on DCP prepared by a Steering Group of field experts (cprn.org/research-cp-dystonia-edition). Participants next submitted their remaining uncertainties about DCP. These were vetted by the Steering Group and consolidated to eliminate redundancy to generate a list of unique uncertainties, which were then prioritized by the participants. The top-prioritized uncertainties were aggregated into themes through iterative consensus-building discussions within the Steering Group. 166 webinar viewers generated 67 unique uncertainties. 29 uncertainties (17 generated by community members) were prioritized higher than their randomly matched pairs. These were coalesced into the following top 10 DCP research themes: (1) develop new treatments; (2) assess rehabilitation, psychological, and environmental management approaches; (3) compare effectiveness of current treatments; (4) improve diagnosis and severity assessments; (5) assess the effect of mixed tone (spasticity and dystonia) in outcomes and approaches; (6) assess predictors of treatment responsiveness; (7) identify pathophysiologic mechanisms; (8) characterize the natural history; (9) determine the best treatments for pain; and (10) increase family awareness. This community-driven research agenda reflects the concerns most important to the community, both in perception and in practice. We therefore encourage future DCP research to center around these themes. Furthermore, noting that community members (not clinicians or researchers) generated the majority of top-prioritized uncertainties, our results highlight the important contributions community members can make to research agendas, even beyond DCP.
Collapse
Affiliation(s)
- Laura A Gilbert
- From the Department of Neurology (L.A.G., B.R.A.), Washington University School of Medicine and St. Louis Children's Hospital, MO; Department of Pediatrics (D.L.F.), University of Toronto and Holland Bloorview Kids Rehabilitation Hospital, Ontario, Canada; Department of Population Health Sciences (P.G., W.K.), University of Utah, Salt Lake City; Departments of Child Health, Neurology, Genetics, and Cellular and Molecular Medicine (M.C.K.), College of Medicine-Phoenix, University of Arizona and Cerebral Palsy and Pediatric Movement Disorders Program, Barrow Neurological Institute, Phoenix Children's Hospital; Department of Neurology (J.W.M.), University of Rochester School of Medicine and Dentistry, NY; and The Cerebral Palsy Research Network (P.G., M.S.), Salt Lake City, UT
| | - Darcy L Fehlings
- From the Department of Neurology (L.A.G., B.R.A.), Washington University School of Medicine and St. Louis Children's Hospital, MO; Department of Pediatrics (D.L.F.), University of Toronto and Holland Bloorview Kids Rehabilitation Hospital, Ontario, Canada; Department of Population Health Sciences (P.G., W.K.), University of Utah, Salt Lake City; Departments of Child Health, Neurology, Genetics, and Cellular and Molecular Medicine (M.C.K.), College of Medicine-Phoenix, University of Arizona and Cerebral Palsy and Pediatric Movement Disorders Program, Barrow Neurological Institute, Phoenix Children's Hospital; Department of Neurology (J.W.M.), University of Rochester School of Medicine and Dentistry, NY; and The Cerebral Palsy Research Network (P.G., M.S.), Salt Lake City, UT
| | - Paul Gross
- From the Department of Neurology (L.A.G., B.R.A.), Washington University School of Medicine and St. Louis Children's Hospital, MO; Department of Pediatrics (D.L.F.), University of Toronto and Holland Bloorview Kids Rehabilitation Hospital, Ontario, Canada; Department of Population Health Sciences (P.G., W.K.), University of Utah, Salt Lake City; Departments of Child Health, Neurology, Genetics, and Cellular and Molecular Medicine (M.C.K.), College of Medicine-Phoenix, University of Arizona and Cerebral Palsy and Pediatric Movement Disorders Program, Barrow Neurological Institute, Phoenix Children's Hospital; Department of Neurology (J.W.M.), University of Rochester School of Medicine and Dentistry, NY; and The Cerebral Palsy Research Network (P.G., M.S.), Salt Lake City, UT
| | - Michael C Kruer
- From the Department of Neurology (L.A.G., B.R.A.), Washington University School of Medicine and St. Louis Children's Hospital, MO; Department of Pediatrics (D.L.F.), University of Toronto and Holland Bloorview Kids Rehabilitation Hospital, Ontario, Canada; Department of Population Health Sciences (P.G., W.K.), University of Utah, Salt Lake City; Departments of Child Health, Neurology, Genetics, and Cellular and Molecular Medicine (M.C.K.), College of Medicine-Phoenix, University of Arizona and Cerebral Palsy and Pediatric Movement Disorders Program, Barrow Neurological Institute, Phoenix Children's Hospital; Department of Neurology (J.W.M.), University of Rochester School of Medicine and Dentistry, NY; and The Cerebral Palsy Research Network (P.G., M.S.), Salt Lake City, UT
| | - Wendy Kwan
- From the Department of Neurology (L.A.G., B.R.A.), Washington University School of Medicine and St. Louis Children's Hospital, MO; Department of Pediatrics (D.L.F.), University of Toronto and Holland Bloorview Kids Rehabilitation Hospital, Ontario, Canada; Department of Population Health Sciences (P.G., W.K.), University of Utah, Salt Lake City; Departments of Child Health, Neurology, Genetics, and Cellular and Molecular Medicine (M.C.K.), College of Medicine-Phoenix, University of Arizona and Cerebral Palsy and Pediatric Movement Disorders Program, Barrow Neurological Institute, Phoenix Children's Hospital; Department of Neurology (J.W.M.), University of Rochester School of Medicine and Dentistry, NY; and The Cerebral Palsy Research Network (P.G., M.S.), Salt Lake City, UT
| | - Jonathan W Mink
- From the Department of Neurology (L.A.G., B.R.A.), Washington University School of Medicine and St. Louis Children's Hospital, MO; Department of Pediatrics (D.L.F.), University of Toronto and Holland Bloorview Kids Rehabilitation Hospital, Ontario, Canada; Department of Population Health Sciences (P.G., W.K.), University of Utah, Salt Lake City; Departments of Child Health, Neurology, Genetics, and Cellular and Molecular Medicine (M.C.K.), College of Medicine-Phoenix, University of Arizona and Cerebral Palsy and Pediatric Movement Disorders Program, Barrow Neurological Institute, Phoenix Children's Hospital; Department of Neurology (J.W.M.), University of Rochester School of Medicine and Dentistry, NY; and The Cerebral Palsy Research Network (P.G., M.S.), Salt Lake City, UT
| | - Michele Shusterman
- From the Department of Neurology (L.A.G., B.R.A.), Washington University School of Medicine and St. Louis Children's Hospital, MO; Department of Pediatrics (D.L.F.), University of Toronto and Holland Bloorview Kids Rehabilitation Hospital, Ontario, Canada; Department of Population Health Sciences (P.G., W.K.), University of Utah, Salt Lake City; Departments of Child Health, Neurology, Genetics, and Cellular and Molecular Medicine (M.C.K.), College of Medicine-Phoenix, University of Arizona and Cerebral Palsy and Pediatric Movement Disorders Program, Barrow Neurological Institute, Phoenix Children's Hospital; Department of Neurology (J.W.M.), University of Rochester School of Medicine and Dentistry, NY; and The Cerebral Palsy Research Network (P.G., M.S.), Salt Lake City, UT
| | - Bhooma R Aravamuthan
- From the Department of Neurology (L.A.G., B.R.A.), Washington University School of Medicine and St. Louis Children's Hospital, MO; Department of Pediatrics (D.L.F.), University of Toronto and Holland Bloorview Kids Rehabilitation Hospital, Ontario, Canada; Department of Population Health Sciences (P.G., W.K.), University of Utah, Salt Lake City; Departments of Child Health, Neurology, Genetics, and Cellular and Molecular Medicine (M.C.K.), College of Medicine-Phoenix, University of Arizona and Cerebral Palsy and Pediatric Movement Disorders Program, Barrow Neurological Institute, Phoenix Children's Hospital; Department of Neurology (J.W.M.), University of Rochester School of Medicine and Dentistry, NY; and The Cerebral Palsy Research Network (P.G., M.S.), Salt Lake City, UT.
| |
Collapse
|
19
|
Xing H, Yokoi F, Walker AL, Torres-Medina R, Liu Y, Li Y. Electrophysiological characterization of the striatal cholinergic interneurons in Dyt1 ΔGAG knock-in mice. DYSTONIA 2022; 1:10557. [PMID: 36329866 PMCID: PMC9629210 DOI: 10.3389/dyst.2022.10557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
DYT1 dystonia is an inherited early-onset movement disorder characterized by sustained muscle contractions causing twisting, repetitive movements, and abnormal postures. Most DYT1 patients have a heterozygous trinucleotide GAG deletion mutation (ΔGAG) in DYT1/TOR1A, coding for torsinA. Dyt1 heterozygous ΔGAG knock-in (KI) mice show motor deficits and reduced striatal dopamine receptor 2 (D2R). Striatal cholinergic interneurons (ChIs) are essential in regulating striatal motor circuits. Multiple dystonia rodent models, including KI mice, show altered ChI firing and modulation. However, due to the errors in assigning KI mice, it is essential to replicate these findings in genetically confirmed KI mice. Here, we found irregular and decreased spontaneous firing frequency in the acute brain slices from Dyt1 KI mice. Quinpirole, a D2R agonist, showed less inhibitory effect on the spontaneous ChI firing in Dyt1 KI mice, suggesting decreased D2R function on the striatal ChIs. On the other hand, a muscarinic receptor agonist, muscarine, inhibited the ChI firing in both wild-type (WT) and Dyt1 KI mice. Trihexyphenidyl, a muscarinic acetylcholine receptor M1 antagonist, had no significant effect on the firing. Moreover, the resting membrane property and functions of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, μ-opioid receptors, and large-conductance calcium-activated potassium (BK) channels were unaffected in Dyt1 KI mice. The results suggest that the irregular and low-frequency firing and decreased D2R function are the main alterations of striatal ChIs in Dyt1 KI mice. These results appear consistent with the reduced dopamine release and high striatal acetylcholine tone in the previous reports.
Collapse
Affiliation(s)
- Hong Xing
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Fumiaki Yokoi
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Ariel Luz Walker
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Rosemarie Torres-Medina
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Yuning Liu
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| | - Yuqing Li
- Norman Fixel Institute of Neurological Diseases and Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610-0236, USA
| |
Collapse
|
20
|
Yellajoshyula D, Rogers AE, Kim AJ, Kim S, Pappas SS, Dauer WT. A pathogenic DYT-THAP1 dystonia mutation causes hypomyelination and loss of YY1 binding. Hum Mol Genet 2022; 31:1096-1104. [PMID: 34686877 PMCID: PMC8976427 DOI: 10.1093/hmg/ddab310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/27/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Dystonia is a disabling disease that manifests as prolonged involuntary twisting movements. DYT-THAP1 is an inherited form of isolated dystonia caused by mutations in THAP1 encoding the transcription factor THAP1. The phe81leu (F81L) missense mutation is representative of a category of poorly understood mutations that do not occur on residues critical for DNA binding. Here, we demonstrate that the F81L mutation (THAP1F81L) impairs THAP1 transcriptional activity and disrupts CNS myelination. Strikingly, THAP1F81L exhibits normal DNA binding but causes a significantly reduced DNA binding of YY1, its transcriptional partner that also has an established role in oligodendrocyte lineage progression. Our results suggest a model of molecular pathogenesis whereby THAP1F81L normally binds DNA but is unable to efficiently organize an active transcription complex.
Collapse
Affiliation(s)
| | - Abigail E Rogers
- Molecular Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Audrey J Kim
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sumin Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Samuel S Pappas
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - William T Dauer
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
21
|
Yellajoshyula D, Pappas SS, Dauer WT. Oligodendrocyte and Extracellular Matrix Contributions to Central Nervous System Motor Function: Implications for Dystonia. Mov Disord 2022; 37:456-463. [PMID: 34989453 PMCID: PMC11152458 DOI: 10.1002/mds.28892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
The quest to elucidate nervous system function and dysfunction in disease has focused largely on neurons and neural circuits. However, fundamental aspects of nervous system development, function, and plasticity are regulated by nonneuronal elements, including glial cells and the extracellular matrix (ECM). The rapid rise of genomics and neuroimaging techniques in recent decades has highlighted neuronal-glial interactions and ECM as a key component of nervous system development, plasticity, and function. Abnormalities of neuronal-glial interactions have been understudied but are increasingly recognized to play a key role in many neurodevelopmental disorders. In this report, we consider the role of myelination and the ECM in the development and function of central nervous system motor circuits and the neurodevelopmental disease dystonia. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Samuel S Pappas
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - William T Dauer
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
22
|
Scarduzio M, Hess EJ, Standaert DG, Eskow Jaunarajs KL. Striatal synaptic dysfunction in dystonia and levodopa-induced dyskinesia. Neurobiol Dis 2022; 166:105650. [DOI: 10.1016/j.nbd.2022.105650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 12/16/2022] Open
|
23
|
Cirnaru MD, Creus-Muncunill J, Nelson S, Lewis TB, Watson J, Ellerby LM, Gonzalez-Alegre P, Ehrlich ME. Striatal Cholinergic Dysregulation after Neonatal Decrease in X-Linked Dystonia Parkinsonism-Related TAF1 Isoforms. Mov Disord 2021; 36:2780-2794. [PMID: 34403156 DOI: 10.1002/mds.28750] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/24/2021] [Accepted: 07/12/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND X-linked dystonia parkinsonism is a generalized, progressive dystonia followed by parkinsonism with onset in adulthood and accompanied by striatal neurodegeneration. Causative mutations are located in a noncoding region of the TATA-box binding protein-associated factor 1 (TAF1) gene and result in aberrant splicing. There are 2 major TAF1 isoforms that may be decreased in symptomatic patients, including the ubiquitously expressed canonical cTAF1 and the neuronal-specific nTAF1. OBJECTIVE The objective of this study was to determine the behavioral and transcriptomic effects of decreased cTAF1 and/or nTAF1 in vivo. METHODS We generated adeno-associated viral (AAV) vectors encoding microRNAs targeting Taf1 in a splice-isoform selective manner. We performed intracerebroventricular viral injections in newborn mice and rats and intrastriatal infusions in 3-week-old rats. The effects of Taf1 knockdown were assayed at 4 months of age with evaluation of motor function, histology, and RNA sequencing of the striatum, followed by its validation. RESULTS We report motor deficits in all cohorts, more pronounced in animals injected at P0, in which we also identified transcriptomic alterations in multiple neuronal pathways, including the cholinergic synapse. In both species, we show a reduced number of striatal cholinergic interneurons and their marker mRNAs after Taf1 knockdown in the newborn. CONCLUSION This study provides novel information regarding the requirement for TAF1 in the postnatal maintenance of striatal cholinergic neurons, the dysfunction of which is involved in other inherited forms of dystonia. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shareen Nelson
- Raymond G. Perelman Center for Cellular & Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Travis B Lewis
- Raymond G. Perelman Center for Cellular & Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jaime Watson
- Raymond G. Perelman Center for Cellular & Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lisa M Ellerby
- Buck Institute for Research on Aging, Novato, California, USA
| | - Pedro Gonzalez-Alegre
- Raymond G. Perelman Center for Cellular & Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
24
|
Frederick NM, Pooler MM, Shah P, Didonna A, Opal P. Pharmacological perturbation reveals deficits in D2 receptor responses in Thap1 null mice. Ann Clin Transl Neurol 2021; 8:2302-2308. [PMID: 34802187 PMCID: PMC8670318 DOI: 10.1002/acn3.51481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 11/17/2022] Open
Abstract
The primary dystonia DYT6 is caused by mutations in the transcription factor Thanatos‐associated protein 1 (THAP1). To understand THAP1’s functions, we generated mice lacking THAP1 in the nervous system. THAP1 loss causes locomotor deficits associated with transcriptional changes. Since many of the genes misregulated involve dopaminergic signaling, we pharmacologically challenged the two striatal canonical dopamine pathways: the direct, regulated by the D1 receptor, and the indirect, regulated by the D2 receptor. We discovered that depleting THAP1 specifically interferes with the D2 receptor responses, pointing to a selective misregulation of the indirect pathway in DYT6 with implications for pathogenesis and treatment.
Collapse
Affiliation(s)
- Natalie M Frederick
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, Illinois, 60208, USA
| | - Morgan M Pooler
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA
| | - Parth Shah
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA
| | - Alessandro Didonna
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, 94158, USA
| | - Puneet Opal
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA.,Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA
| |
Collapse
|
25
|
Selective Manipulation of G-Protein γ 7 Subunit in Mice Provides New Insights into Striatal Control of Motor Behavior. J Neurosci 2021; 41:9065-9081. [PMID: 34544837 DOI: 10.1523/jneurosci.1211-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/26/2021] [Accepted: 09/11/2021] [Indexed: 01/15/2023] Open
Abstract
Stimulatory coupling of dopamine D1 (D1R) and adenosine A2A receptors (A2AR) to adenylyl cyclase within the striatum is mediated through a specific Gαolfβ2γ7 heterotrimer to ultimately modulate motor behaviors. To dissect the individual roles of the Gαolfβ2γ7 heterotrimer in different populations of medium spiny neurons (MSNs), we produced and characterized conditional mouse models, in which the Gng7 gene was deleted in either the D1R- or A2AR/D2R-expressing MSNs. We show that conditional loss of γ7 disrupts the cell type-specific assembly of the Gαolfβ2γ7 heterotrimer, thereby identifying its circumscribed roles acting downstream of either the D1Rs or A2ARs in coordinating motor behaviors, including in vivo responses to psychostimulants. We reveal that Gαolfβ2γ7/cAMP signal in D1R-MSNs does not impact spontaneous and amphetamine-induced locomotor behaviors in male and female mice, while its loss in A2AR/D2R-MSNs results in a hyperlocomotor phenotype and enhanced locomotor response to amphetamine. Additionally, Gαolfβ2γ7/cAMP signal in either D1R- or A2AR/D2R-expressing MSNs is not required for the activation of PKA signaling by amphetamine. Finally, we show that Gαolfβ2γ7 signaling acting downstream of D1Rs is selectively implicated in the acute locomotor-enhancing effects of morphine. Collectively, these results support the general notion that receptors use specific Gαβγ proteins to direct the fidelity of downstream signaling pathways and to elicit a diverse repertoire of cellular functions. Specifically, these findings highlight the critical role for the γ7 protein in determining the cellular level, and hence, the function of the Gαolfβ2γ7 heterotrimer in several disease states associated with dysfunctional striatal signaling.SIGNIFICANCE STATEMENT Dysfunction or imbalance of cAMP signaling in the striatum has been linked to several neurologic and neuropsychiatric disorders, including Parkinson's disease, dystonia, schizophrenia, and drug addiction. By genetically targeting the γ7 subunit in distinct striatal neuronal subpopulations in mice, we demonstrate that the formation and function of the Gαolfβ2γ7 heterotrimer, which represents the rate-limiting step for cAMP production in the striatum, is selectively disrupted. Furthermore, we reveal cell type-specific roles for Gαolfβ2γ7-mediated cAMP production in the control of spontaneous locomotion as well as behavioral and molecular responses to psychostimulants. Our findings identify the γ7 protein as a novel therapeutic target for disease states associated with dysfunctional striatal cAMP signaling.
Collapse
|
26
|
Wilkes BJ, DeSimone JC, Liu Y, Chu WT, Coombes SA, Li Y, Vaillancourt DE. Cell-specific effects of Dyt1 knock-out on sensory processing, network-level connectivity, and motor deficits. Exp Neurol 2021; 343:113783. [PMID: 34119482 PMCID: PMC8324325 DOI: 10.1016/j.expneurol.2021.113783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 11/19/2022]
Abstract
DYT1 dystonia is a debilitating movement disorder characterized by repetitive, unintentional movements and postures. The disorder has been linked to mutation of the TOR1A/DYT1 gene encoding torsinA. Convergent evidence from studies in humans and animal models suggest that striatal medium spiny neurons and cholinergic neurons are important in DYT1 dystonia. What is not known is how torsinA dysfunction in these specific cell types contributes to the pathophysiology of DYT1 dystonia. In this study we sought to determine whether torsinA dysfunction in cholinergic neurons alone is sufficient to generate the sensorimotor dysfunction and brain changes associated with dystonia, or if torsinA dysfunction in a broader subset of cell types is needed. We generated two genetically modified mouse models, one with selective Dyt1 knock-out from dopamine-2 receptor expressing neurons (D2KO) and one where only cholinergic neurons are impacted (Ch2KO). We assessed motor deficits and performed in vivo 11.1 T functional MRI to assess sensory-evoked brain activation and connectivity, along with diffusion MRI to assess brain microstructure. We found that D2KO mice showed greater impairment than Ch2KO mice, including reduced sensory-evoked brain activity in key regions of the sensorimotor network, and altered functional connectivity of the striatum that correlated with motor deficits. These findings suggest that (1) the added impact of torsinA dysfunction in medium spiny and dopaminergic neurons of the basal ganglia generate more profound deficits than the dysfunction of cholinergic neurons alone, and (2) that sensory network impairments are linked to motor deficits in DYT1 dystonia.
Collapse
Affiliation(s)
- B J Wilkes
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA.
| | - J C DeSimone
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Y Liu
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - W T Chu
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - S A Coombes
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Y Li
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - D E Vaillancourt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Department of Neurology, University of Florida, Gainesville, FL, USA; Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
27
|
Caffall ZF, Wilkes BJ, Hernández-Martinez R, Rittiner JE, Fox JT, Wan KK, Shipman MK, Titus SA, Zhang YQ, Patnaik S, Hall MD, Boxer MB, Shen M, Li Z, Vaillancourt DE, Calakos N. The HIV protease inhibitor, ritonavir, corrects diverse brain phenotypes across development in mouse model of DYT-TOR1A dystonia. Sci Transl Med 2021; 13:13/607/eabd3904. [PMID: 34408078 DOI: 10.1126/scitranslmed.abd3904] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/14/2020] [Accepted: 06/03/2021] [Indexed: 12/22/2022]
Abstract
Dystonias are a group of chronic movement-disabling disorders for which highly effective oral medications or disease-modifying therapies are lacking. The most effective treatments require invasive procedures such as deep brain stimulation. In this study, we used a high-throughput assay based on a monogenic form of dystonia, DYT1 (DYT-TOR1A), to screen a library of compounds approved for use in humans, the NCATS Pharmaceutical Collection (NPC; 2816 compounds), and identify drugs able to correct mislocalization of the disease-causing protein variant, ∆E302/3 hTorsinA. The HIV protease inhibitor, ritonavir, was among 18 compounds found to normalize hTorsinA mislocalization. Using a DYT1 knock-in mouse model to test efficacy on brain pathologies, we found that ritonavir restored multiple brain abnormalities across development. Ritonavir acutely corrected striatal cholinergic interneuron physiology in the mature brain and yielded sustained correction of diffusion tensor magnetic resonance imaging signals when delivered during a discrete early developmental window. Mechanistically, we found that, across the family of HIV protease inhibitors, efficacy correlated with integrated stress response activation. These preclinical results identify ritonavir as a drug candidate for dystonia with disease-modifying potential.
Collapse
Affiliation(s)
- Zachary F Caffall
- Department of Neurology, Duke University Medical Center, Durham, NC 27715, USA
| | - Bradley J Wilkes
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | | | - Joseph E Rittiner
- Department of Neurology, Duke University Medical Center, Durham, NC 27715, USA
| | - Jennifer T Fox
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Kanny K Wan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Miranda K Shipman
- Department of Neurology, Duke University Medical Center, Durham, NC 27715, USA
| | - Steven A Titus
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Ya-Qin Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Samarjit Patnaik
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Matthew B Boxer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Zhuyin Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - David E Vaillancourt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA.,Department of Neurology, Fixel Institute for Neurological Diseases, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Nicole Calakos
- Department of Neurology, Duke University Medical Center, Durham, NC 27715, USA. .,Department of Neurobiology, Duke University Medical Center, Durham, NC 27715, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27715, USA.,Duke Institute for Brain Sciences, Duke University, Durham, NC 27715, USA
| |
Collapse
|
28
|
Behavioral and neurochemical studies of inherited manganese-induced dystonia-parkinsonism in Slc39a14-knockout mice. Neurobiol Dis 2021; 158:105467. [PMID: 34358615 DOI: 10.1016/j.nbd.2021.105467] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/14/2021] [Accepted: 08/02/2021] [Indexed: 11/21/2022] Open
Abstract
Inherited autosomal recessive mutations of the manganese (Mn) transporter gene SLC39A14 in humans, results in elevated blood and brain Mn concentrations and childhood-onset dystonia-parkinsonism. The pathophysiology of this disease is unknown, but the nigrostriatal dopaminergic system of the basal ganglia has been implicated. Here, we describe pathophysiological studies in Slc39a14-knockout (KO) mice as a preclinical model of dystonia-parkinsonism in SLC39A14 mutation carriers. Blood and brain metal concentrations in Slc39a14-KO mice exhibited a pattern similar to the human disease with highly elevated Mn concentrations. We observed an early-onset backward-walking behavior at postnatal day (PN) 21 which was also noted in PN60 Slc39a14-KO mice as well as dystonia-like movements. Locomotor activity and motor coordination were also impaired in Slc39a14-KO relative to wildtype (WT) mice. From a neurochemical perspective, striatal dopamine (DA) and metabolite concentrations and their ratio in Slc39a14-KO mice did not differ from WT. Striatal tyrosine hydroxylase (TH) immunohistochemistry did not change in Slc39a14-KO mice relative to WT. Unbiased stereological cell quantification of TH-positive and Nissl-stained estimated neuron number, neuron density, and soma volume in the substantia nigra pars compacta (SNc) was the same in Slc39a14-KO mice as in WT. However, we measured a marked inhibition (85-90%) of potassium-stimulated DA release in the striatum of Slc39a14-KO mice relative to WT. Our findings indicate that the dystonia-parkinsonism observed in this genetic animal model of the human disease is associated with a dysfunctional but structurally intact nigrostriatal dopaminergic system. The presynaptic deficit in DA release is unlikely to explain the totality of the behavioral phenotype and points to the involvement of other neuronal systems and brain regions in the pathophysiology of the disease.
Collapse
|
29
|
Briscione MA, Dinasarapu AR, Bagchi P, Donsante Y, Roman KM, Downs AM, Fan X, Hoehner J, Jinnah HA, Hess EJ. Differential expression of striatal proteins in a mouse model of DOPA-responsive dystonia reveals shared mechanisms among dystonic disorders. Mol Genet Metab 2021; 133:352-361. [PMID: 34092491 PMCID: PMC8292208 DOI: 10.1016/j.ymgme.2021.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 11/23/2022]
Abstract
Dystonia is characterized by involuntary muscle contractions that cause debilitating twisting movements and postures. Although dysfunction of the basal ganglia, a brain region that mediates movement, is implicated in many forms of dystonia, the underlying mechanisms are unclear. The inherited metabolic disorder DOPA-responsive dystonia is considered a prototype for understanding basal ganglia dysfunction in dystonia because it is caused by mutations in genes necessary for the synthesis of the neurotransmitter dopamine, which mediates the activity of the basal ganglia. Therefore, to reveal abnormal striatal cellular processes and pathways implicated in dystonia, we used an unbiased proteomic approach in a knockin mouse model of DOPA-responsive dystonia, a model in which the striatum is known to play a central role in the expression of dystonia. Fifty-seven of the 1805 proteins identified were differentially regulated in DOPA-responsive dystonia mice compared to control mice. Most differentially regulated proteins were associated with gene ontology terms that implicated either mitochondrial or synaptic dysfunction whereby proteins associated with mitochondrial function were generally over-represented and proteins associated with synaptic function were largely under-represented. Remarkably, nearly 20% of the differentially regulated striatal proteins identified in our screen are associated with pathogenic variants that cause inherited disorders with dystonia as a sign in humans suggesting shared mechanisms across many different forms of dystonia.
Collapse
Affiliation(s)
- Maria A Briscione
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | | | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory University, Atlanta, GA, USA
| | - Yuping Donsante
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Kaitlyn M Roman
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Anthony M Downs
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Xueliang Fan
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Jessica Hoehner
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - H A Jinnah
- Department of Human Genetics, Emory University, Atlanta, GA, USA; Department of Neurology, Emory University, Atlanta, GA, USA; Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Ellen J Hess
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA; Department of Neurology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
30
|
Tassone A, Martella G, Meringolo M, Vanni V, Sciamanna G, Ponterio G, Imbriani P, Bonsi P, Pisani A. Vesicular Acetylcholine Transporter Alters Cholinergic Tone and Synaptic Plasticity in DYT1 Dystonia. Mov Disord 2021; 36:2768-2779. [PMID: 34173686 PMCID: PMC9291835 DOI: 10.1002/mds.28698] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 11/17/2022] Open
Abstract
Background Acetylcholine‐mediated transmission plays a central role in the impairment of corticostriatal synaptic activity and plasticity in multiple DYT1 mouse models. However, the nature of such alteration remains unclear. Objective The aim of the present work was to characterize the mechanistic basis of cholinergic dysfunction in DYT1 dystonia to identify potential targets for pharmacological intervention. Methods We utilized electrophysiology recordings, immunohistochemistry, enzymatic activity assays, and Western blotting techniques to analyze in detail the cholinergic machinery in the dorsal striatum of the Tor1a+/− mouse model of DYT1 dystonia. Results We found a significant increase in the vesicular acetylcholine transporter (VAChT) protein level, the protein responsible for loading acetylcholine (ACh) from the cytosol into synaptic vesicles, which indicates an altered cholinergic tone. Accordingly, in Tor1a+/− mice we measured a robust elevation in basal ACh content coupled to a compensatory enhancement of acetylcholinesterase (AChE) enzymatic activity. Moreover, pharmacological activation of dopamine D2 receptors, which is expected to reduce ACh levels, caused an abnormal elevation in its content, as compared to controls. Patch‐clamp recordings revealed a reduced effect of AChE inhibitors on cholinergic interneuron excitability, whereas muscarinic autoreceptor function was preserved. Finally, we tested the hypothesis that blockade of VAChT could restore corticostriatal long‐term synaptic plasticity deficits. Vesamicol, a selective VAChT inhibitor, rescued a normal expression of synaptic plasticity. Conclusions Overall, our findings indicate that VAChT is a key player in the alterations of striatal plasticity and a novel target to normalize cholinergic dysfunction observed in DYT1 dystonia. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Annalisa Tassone
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Meringolo
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Valentina Vanni
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppe Sciamanna
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giulia Ponterio
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Imbriani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
31
|
Danielsson K, Lagström O, Ericson M, Söderpalm B, Adermark L. Subregion-specific effects on striatal neurotransmission and dopamine-signaling by acute and repeated amphetamine exposure. Neuropharmacology 2021; 194:108638. [PMID: 34116108 DOI: 10.1016/j.neuropharm.2021.108638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 11/29/2022]
Abstract
Repeated administration of psychostimulants, such as amphetamine, is associated with a progressive increased sensitivity to some of the drug's effects, but tolerance towards others. We hypothesized that these adaptations in part could be linked to differential effects by amphetamine on dopaminergic signaling in striatal subregions. To test this theory, acute and long-lasting changes in dopaminergic neurotransmission were assessed in the nucleus accumbens (nAc) and the dorsomedial striatum (DMS) following amphetamine exposure in Wistar rats. By means of in vivo microdialysis, dopamine release induced by local administration of amphetamine was monitored in nAc and DMS of amphetamine naïve rats, and in rats subjected to five days of systemic amphetamine administration (2.0 mg/kg/day) followed by two weeks of withdrawal. In parallel, ex vivo electrophysiology was conducted to outline the effect of acute and repeated amphetamine exposure on striatal neurotransmission. The data shows that amphetamine increases dopamine in a concentration-dependent and subregion-specific manner. Furthermore, repeated administration of amphetamine followed by abstinence resulted in a selective decrease in baseline dopamine in the nAc, and a potentiation of the relative dopamine elevation after systemic amphetamine in the same area. Ex vivo electrophysiology demonstrated decreased excitatory neurotransmission in brain slices from amphetamine-treated animals, and a nAc selective shift in the responsiveness to the dopamine D2-receptor agonist quinpirole. These selective effects on dopamine signaling seen in striatal subregions after repeated drug exposure may partially explain why tolerance develops to the rewarding effects, but not towards the psychosis inducing properties of amphetamine.
Collapse
Affiliation(s)
- Klara Danielsson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Oona Lagström
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Mia Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Bo Söderpalm
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden; Beroendekliniken, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Louise Adermark
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden; Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy University of Gothenburg, Sweden.
| |
Collapse
|
32
|
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
|
33
|
Poppi LA, Ho-Nguyen KT, Shi A, Daut CT, Tischfield MA. Recurrent Implication of Striatal Cholinergic Interneurons in a Range of Neurodevelopmental, Neurodegenerative, and Neuropsychiatric Disorders. Cells 2021; 10:907. [PMID: 33920757 PMCID: PMC8071147 DOI: 10.3390/cells10040907] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/03/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Cholinergic interneurons are "gatekeepers" for striatal circuitry and play pivotal roles in attention, goal-directed actions, habit formation, and behavioral flexibility. Accordingly, perturbations to striatal cholinergic interneurons have been associated with many neurodevelopmental, neurodegenerative, and neuropsychiatric disorders. The role of acetylcholine in many of these disorders is well known, but the use of drugs targeting cholinergic systems fell out of favor due to adverse side effects and the introduction of other broadly acting compounds. However, in response to recent findings, re-examining the mechanisms of cholinergic interneuron dysfunction may reveal key insights into underlying pathogeneses. Here, we provide an update on striatal cholinergic interneuron function, connectivity, and their putative involvement in several disorders. In doing so, we aim to spotlight recurring physiological themes, circuits, and mechanisms that can be investigated in future studies using new tools and approaches.
Collapse
Affiliation(s)
- Lauren A. Poppi
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA;
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Tourette International Collaborative (TIC) Genetics Study, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Khue Tu Ho-Nguyen
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Anna Shi
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Cynthia T. Daut
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Max A. Tischfield
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; (K.T.H.-N.); (A.S.); (C.T.D.)
- Tourette International Collaborative (TIC) Genetics Study, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
34
|
Alteration of the cholinergic system and motor deficits in cholinergic neuron-specific Dyt1 knockout mice. Neurobiol Dis 2021; 154:105342. [PMID: 33757902 DOI: 10.1016/j.nbd.2021.105342] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/11/2021] [Accepted: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Dystonia is a neurological movement disorder characterized by sustained or intermittent muscle contractions, repetitive movement, and sometimes abnormal postures. DYT1 dystonia is one of the most common genetic dystonias, and most patients carry heterozygous DYT1 ∆GAG mutations causing a loss of a glutamic acid of the protein torsinA. Patients can be treated with anticholinergics, such as trihexyphenidyl, suggesting an abnormal cholinergic state. Early work on the cell-autonomous effects of Dyt1 deletion with ChI-specific Dyt1 conditional knockout mice (Dyt1 Ch1KO) revealed abnormal electrophysiological responses of striatal ChIs to muscarine and quinpirole, motor deficits, and no changes in the number or size of the ChIs. However, the Chat-cre line that was used to derive Dyt1 Ch1KO mice contained a neomycin cassette and was reported to have ectopic cre-mediated recombination. In this study, we generated a Dyt1 Ch2KO mouse line by removing the neomycin cassette in Dyt1 Ch1KO mice. The Dyt1 Ch2KO mice showed abnormal paw clenching behavior, motor coordination and balance deficits, impaired motor learning, reduced striatal choline acetyltransferase protein level, and a reduced number of striatal ChIs. Furthermore, the mutant striatal ChIs had a normal muscarinic inhibitory function, impaired quinpirole-mediated inhibition, and altered current density. Our findings demonstrate a cell-autonomous effect of Dyt1 deletion on the striatal ChIs and a critical role for the striatal ChIs and corticostriatal pathway in the pathogenesis of DYT1 dystonia.
Collapse
|
35
|
Melis C, Beauvais G, Muntean BS, Cirnaru MD, Otrimski G, Creus-Muncunill J, Martemyanov KA, Gonzalez-Alegre P, Ehrlich ME. Striatal Dopamine Induced ERK Phosphorylation Is Altered in Mouse Models of Monogenic Dystonia. Mov Disord 2021; 36:1147-1157. [PMID: 33458877 DOI: 10.1002/mds.28476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Similar to some monogenic forms of dystonia, levodopa-induced dyskinesia is a hyperkinetic movement disorder with abnormal nigrostriatal dopaminergic neurotransmission. Molecularly, it is characterized by hyper-induction of phosphorylation of extracellular signal-related kinase in response to dopamine in medium spiny neurons of the direct pathway. OBJECTIVES The objective of this study was to determine if mouse models of monogenic dystonia exhibit molecular features of levodopa-induced dyskinesia. METHODS Western blotting and quantitative immunofluorescence was used to assay baseline and/or dopamine-induced levels of the phosphorylated kinase in the striatum in mouse models of DYT1, DYT6, and DYT25 expressing a reporter in dopamine D1 receptor-expressing projection neurons. Cyclic adenosine monophosphate (cAMP) immunoassay and adenylyl cyclase activity assays were also performed. RESULTS In DYT1 and DYT6 models, blocking dopamine reuptake with cocaine leads to enhanced extracellular signal-related kinase phosphorylation in dorsomedial striatal medium spiny neurons in the direct pathway, which is abolished by pretreatment with the N-methyl-d-aspartate antagonist MK-801. Phosphorylation is decreased in a model of DYT25. Levels of basal and stimulated cAMP and adenylyl cyclase activity were normal in the DYT1 and DYT6 mice and decreased in the DYT25 mice. Oxotremorine induced increased abnormal movements in the DYT1 knock-in mice. CONCLUSIONS The increased dopamine induction of extracellular signal-related kinase phosphorylation in 2 genetic types of dystonia, similar to what occurs in levodopa-induced dyskinesia, and its decrease in a third, suggests that abnormal signal transduction in response to dopamine in the postsynaptic nigrostriatal pathway might be a point of convergence for dystonia and other hyperkinetic movement disorders, potentially offering common therapeutic targets. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chiara Melis
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Genevieve Beauvais
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brian S Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Maria-Daniela Cirnaru
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Garrett Otrimski
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | - Pedro Gonzalez-Alegre
- Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurology, The University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
36
|
Li J, Liang CC, Pappas SS, Dauer WT. TorsinB overexpression prevents abnormal twisting in DYT1 dystonia mouse models. eLife 2020; 9:e54285. [PMID: 32202496 PMCID: PMC7141835 DOI: 10.7554/elife.54285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/23/2020] [Indexed: 12/13/2022] Open
Abstract
Genetic redundancy can be exploited to identify therapeutic targets for inherited disorders. We explored this possibility in DYT1 dystonia, a neurodevelopmental movement disorder caused by a loss-of-function (LOF) mutation in the TOR1A gene encoding torsinA. Prior work demonstrates that torsinA and its paralog torsinB have conserved functions at the nuclear envelope. This work established that low neuronal levels of torsinB dictate the neuronal selective phenotype of nuclear membrane budding. Here, we examined whether torsinB expression levels impact the onset or severity of abnormal movements or neuropathological features in DYT1 mouse models. We demonstrate that torsinB levels bidirectionally regulate these phenotypes. Reducing torsinB levels causes a dose-dependent worsening whereas torsinB overexpression rescues torsinA LOF-mediated abnormal movements and neurodegeneration. These findings identify torsinB as a potent modifier of torsinA LOF phenotypes and suggest that augmentation of torsinB expression may retard or prevent symptom development in DYT1 dystonia.
Collapse
Affiliation(s)
- Jay Li
- Medical Scientist Training Program, University of MichiganAnn ArborUnited States
- Cellular and Molecular Biology Graduate Program, University of MichiganAnn ArborUnited States
| | - Chun-Chi Liang
- Department of Neurology, University of MichiganAnn ArborUnited States
| | - Samuel S Pappas
- Peter O’Donnell Jr. Brain Institute, Departments of Neuroscience and Neurology & Neurotherapeutics, University of Texas SouthwesternDallasUnited States
| | - William T Dauer
- Department of Neurology, University of MichiganAnn ArborUnited States
- Peter O’Donnell Jr. Brain Institute, Departments of Neuroscience and Neurology & Neurotherapeutics, University of Texas SouthwesternDallasUnited States
| |
Collapse
|