1
|
Pollini L, Pettenuzzo I, Tijssen MAJ, Koens LH, De Koning TJ, Leuzzi V, Eggink H. Eye movement disorders in genetic dystonia syndromes: A literature overview. Parkinsonism Relat Disord 2025:107325. [PMID: 39966058 DOI: 10.1016/j.parkreldis.2025.107325] [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: 11/27/2024] [Revised: 01/17/2025] [Accepted: 02/09/2025] [Indexed: 02/20/2025]
Abstract
With the growing possibilities in genetic testing, the number of genetic disorders associated with dystonia has constantly increased over the last few years. Accurate phenotyping is crucial to guide and interpret genetic analyses in the search for an etiological diagnosis. Although eye movements examination has proven a valuable tool in the assessment of patients with inherited movement disorders such as ataxia or parkinsonism, less is known about the association between eye movement disorders and genetic dystonia. This study aimed to summarize the most frequent eye movement disorders in monogenetic forms of dystonia as classified by the Movement Disorders Society (MDS). More than sixty genetic disorders causing dystonia were repeatedly associated with eye movement disorders. Among these, 24 are classified as DYT genes, 22 were classified by MDS as having another prominent movement disorder, and 19 are genetic disorders that manifest with dystonia but are not included in the MDS classification. Six different eye movement disorders have consistently been reported (saccadic slowing and supranuclear gaze palsy, saccadic initiation failure and oculomotor apraxia, saccadic dysmetria, oculogyric crisis, nystagmus and ophthalmoplegia). The phenotypic association of each disorder with monogenic dystonic diseases, as well as the possible underlying pathophysiological mechanisms, is described here. Our findings suggest that eye movement disorders, along with the movement phenotype, may help delineate subgroups of dystonia by reflecting disruptions in specific brain networks. Therefore, eye movement examination is a crucial part of the neurological evaluation, providing valuable insights into patients with inherited forms of dystonia.
Collapse
Affiliation(s)
- Luca Pollini
- Department of Human Neuroscience, Sapienza University of Rome, 00185, Rome, Italy; Expertise Centre Movement Disorders Groningen, University Medical Centre Groningen (UMCG), Groningen, the Netherlands
| | - Ilaria Pettenuzzo
- Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy; IRCCS Istituto delle Scienze Neurologiche di Bologna, U.O.C. Neuropsichiatria dell'età pediatrica, Bologna, Italy; Expertise Centre Movement Disorders Groningen, University Medical Centre Groningen (UMCG), Groningen, the Netherlands
| | - Marina A J Tijssen
- Department of Neurology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, the Netherlands; Expertise Centre Movement Disorders Groningen, University Medical Centre Groningen (UMCG), Groningen, the Netherlands
| | - Lisette H Koens
- Department of Neurology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, the Netherlands; Expertise Centre Movement Disorders Groningen, University Medical Centre Groningen (UMCG), Groningen, the Netherlands; Department of Neurology and Clinical Neurophysiology, Martini Ziekenhuis, Groningen, the Netherlands
| | - Tom J De Koning
- Expertise Centre Movement Disorders Groningen, University Medical Centre Groningen (UMCG), Groningen, the Netherlands; Pediatrics, department of Clinical Sciences, Lund University, Sweden
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, Sapienza University of Rome, 00185, Rome, Italy
| | - Hendriekje Eggink
- Department of Neurology, University of Groningen, University Medical Centre Groningen (UMCG), Groningen, the Netherlands; Expertise Centre Movement Disorders Groningen, University Medical Centre Groningen (UMCG), Groningen, the Netherlands.
| |
Collapse
|
2
|
Hassan MT, Radhakrishnan SK, Sharma S, Lytton J. Cellular and subcellular distribution of the K +-dependent Na +/Ca 2+-exchanger subtype 4, NCKX4, in mouse brain. Neuroscience 2025; 569:210-230. [PMID: 39923981 DOI: 10.1016/j.neuroscience.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
SLC24A4, the gene encoding the K+-dependent Na+/Ca2+-exchanger, NCKX4, is widely expressed in the brain and involved in many neuronal functions. Here we use immunofluorescent staining and electron microscopy to map the expression of the NCKX4 protein across the mouse brain. Our data show that NCKX4 expression is higher in forebrain with particularly intense staining in specific brain regions, including olfactory bulb, ventral pallidum, globus pallidus, hippocampal mossy fibers, substantia nigra, inferior olive and choroid plexus. These areas are closely associated with crucial functions such as learning and memory, reward processing, motor activity, and cerebrospinal fluid production. Co-immunostaining of NCKX4 with marker proteins and immuno-electron microscopy demonstrate that neuronal NCKX4 is not expressed in cell bodies but is confined to distal neuronal processes with preferential expression on the cell surface. In contrast, NCKX4 expression in epithelial cells of the choroid plexus is found exclusively at intracellular sites. We also compared NCKX4 to two other Na+/Ca2+-exchangers, NCKX2 and NCX1. NCKX4 is the only exchanger expressed in choroid plexus and hippocampal mossy fibers. In the substantia nigra, NCKX4 and NCKX2 show striking overlap while NCX1 is absent. In many other brain regions all three exchangers are present. These data suggest both distinct and redundant roles for these exchangers in different brain regions. This novel information provides valuable insight into the mechanisms underlying the contribution of NCKX4 to various physiological processes associated with different brain regions and suggests the existence of undiscovered roles for NCKX4 in the brain.
Collapse
Affiliation(s)
- Mohamed Tarek Hassan
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Sarvan Kumar Radhakrishnan
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Sunita Sharma
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Jonathan Lytton
- Department of Biochemistry & Molecular Biology, Libin Cardiovascular Institute and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada.
| |
Collapse
|
3
|
Tolve M, Tutas J, Özer-Yildiz E, Klein I, Petzold A, Fritz VJ, Overhoff M, Silverman Q, Koletsou E, Liebsch F, Schwarz G, Korotkova T, Valtcheva S, Gatto G, Kononenko NL. The endocytic adaptor AP-2 maintains Purkinje cell function by balancing cerebellar parallel and climbing fiber synapses. Cell Rep 2025; 44:115256. [PMID: 39918958 DOI: 10.1016/j.celrep.2025.115256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/02/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
The loss of cerebellar Purkinje cells is a hallmark of neurodegenerative movement disorders, but the mechanisms remain enigmatic. We show that endocytic adaptor protein complex 2 (AP-2) is crucial for Purkinje cell survival. Using mouse genetics, viral tracing, calcium imaging, and kinematic analysis, we demonstrate that loss of the AP-2 μ subunit in Purkinje cells leads to early-onset ataxia and progressive degeneration. Synaptic dysfunction, marked by an overrepresentation of parallel fibers (PFs) over climbing fibers (CFs), precedes Purkinje cell loss. Mechanistically, AP-2 interacts with the PF-enriched protein GRID2IP, and its loss triggers GRID2IP degradation and glutamate δ2 receptor (GLURδ2) accumulation, leading to an excess of PFs while CFs are reduced. The overrepresentation of PFs increases Purkinje cell network activity, which is mitigated by enhancing glutamate clearance with ceftriaxone. These findings highlight the role of AP-2 in regulating GRID2IP levels in Purkinje cells to maintain PF-CF synaptic balance and prevent motor dysfunction.
Collapse
Affiliation(s)
- Marianna Tolve
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Janine Tutas
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ebru Özer-Yildiz
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ines Klein
- Neurology Department, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Anne Petzold
- Institute for Systems Physiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Veronika J Fritz
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Melina Overhoff
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Quinn Silverman
- Neurology Department, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Ellie Koletsou
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Filip Liebsch
- Institute of Biochemistry, Department of Chemistry, University of Cologne, Cologne, Germany
| | - Guenter Schwarz
- Institute of Biochemistry, Department of Chemistry, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Tatiana Korotkova
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Institute for Systems Physiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Silvana Valtcheva
- Institute for Systems Physiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Graziana Gatto
- Neurology Department, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Natalia L Kononenko
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Institute of Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
4
|
Widner J, Faust PL, Louis ED, Fujita H. Axonal pathology differentially affects human Purkinje cell subtypes in the essential tremor cerebellum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.26.633063. [PMID: 39974874 PMCID: PMC11838201 DOI: 10.1101/2025.01.26.633063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The cerebellar cortex is organized into discrete regions populated by molecularly distinct Purkinje cells (PCs), the sole cortical output neurons. While studies in animal models have shown that PC subtypes differ in their vulnerability to disease, our understanding of human PC subtype and vulnerability remains limited. Here, we demonstrate that human cerebellar regions specialized for motor vs cognitive functions (lobule HV vs Crus I) contain distinct PC populations characterized by specific molecular and anatomical features, which show selective vulnerability in essential tremor (ET), a cerebellar degenerative disorder. Using a known PC subtype marker, neurofilament heavy chain (NEFH), we found that motor lobule HV contains PCs with high NEFH expression, while cognitive lobule Crus I contains PCs with low NEFH expression in post-mortem samples from healthy controls. In the same cerebella, PC axons in lobule HV were 2.2-fold thicker than those in Crus I. Across lobules, axon caliber positively correlated with NEFH expression. In ET cerebella, we identified motor lobule-specific PC axon pathology with a 1.5-fold reduction in caliber and increased axon variability in lobule HV, while Crus I axons were unaffected. Tremor severity and duration in ET correlated with axon diameter variability selectively in lobule HV PCs. Given that axonal caliber is a major determinant of neural signaling capacity, our results (1) suggest that disrupted cerebellar corticonuclear signaling is occurring in ET, (2) provide evidence of region-specific PC subtypes in the human cerebellum and offer insight into how selective PC vulnerability may contribute to the pathophysiology of cerebellar degeneration. Significance Statement The cerebellar cortex has a uniform laminar architecture but contains heterogeneous cell types. Purkinje cells (PCs), the sole output cells of the cerebellar cortex, include subtypes whose lobular distribution is thought to underlie functional segmentation and patterned degeneration of the cerebellum in animal models. However, human PC subtypes and their disease vulnerability remain unknown. Here, we establish the existence of human PC subtypes that appear conserved across mammalian species using both marker expression and axonal thickness. Consistent with its phenotype, we demonstrate differential PC vulnerability to degeneration in essential tremor cerebella, where motor function- mediating PC subtypes display significant axonal thinning, while axons of cognitive function-mediating PC subtypes are spared. These findings advance Purkinje cell type- oriented research in cerebellar disorders.
Collapse
|
5
|
Shih LC. Emerging Deep Brain Stimulation Targets in the Cerebellum for Tremor. CEREBELLUM (LONDON, ENGLAND) 2025; 24:38. [PMID: 39869292 DOI: 10.1007/s12311-025-01789-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/10/2025] [Indexed: 01/28/2025]
Abstract
Deep brain stimulation (DBS) for essential tremor is remarkably effective, leading to over 80% reduction in standardized tremor ratings. However, for certain types of tremor, such as those accompanied by ataxia or dystonia, conventional DBS targets have shown poor efficacy. Various rationales for using cerebellar DBS stimulation to treat tremor have been advanced, but the varied approaches leave many questions unanswered: which anatomic target, stimulation settings, and indications seem most promising for this emerging approach. This article reviews the clinical experience published to date and explores some of the pre-clinical and human physiology data that might support a role for further systematic investigation of cerebellar DBS for clinical use. Four tremor disorders to date have been targeted with cerebellar DBS in humans: essential tremor, post-stroke tremor, dystonic tremor, and tremor associated with degenerative cerebellar ataxias, like spinocerebellar ataxia type 3. The dentate nucleus is the most frequently chosen target, but key stereotactic and imaging details are missing from many of the case reports. Interestingly, consensus on laterality has not been definitively established as there are conflicting models of the hypothesized mechanism of action of DBS of the dentate nuclei, and conflicting reports of benefit on the tremor ipsilateral to and contralateral toto the affected limb. Several points are highlighted, including the prediction from in vivo preclinical physiology studies and interventional studies, the remaining uncertainty regarding the preferred laterality of targets, and the lack of clear prioritization of tremor etiologies to be targeted in future rigorously designed interventional studies (e.g., preferably repeated n-of-1 or sham-controlled studies involving more than one patient).
Collapse
Affiliation(s)
- Ludy C Shih
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Huang X, Fu X, Wu J, Cheng X, Hong X, Li Z, Zheng L, Liu Q, Chen S, Tang B, Zhao Y, Liu X, Li X, Liu X, Zhou Z, Wu L, Fang K, Zhong P, Zhang M, Luan X, Tian W, Tong X, Cao L. Heterozygous KCNJ10 Variants Affecting Kir4.1 Channel Cause Paroxysmal Kinesigenic Dyskinesia. Mov Disord 2024; 39:2199-2210. [PMID: 39367724 DOI: 10.1002/mds.30025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 08/15/2024] [Accepted: 09/12/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND More than 60% of paroxysmal kinesigenic dyskinesia (PKD) cases are of uncertain variants. OBJECTIVE The aim was to elucidate novel genetic contribution to PKD. METHODS A total of 476 probands with uncertain genetic causes were enrolled for whole-exome sequencing. A method of case-control analysis was applied to identify the candidate genes. Whole-cell patch-clamp recording was applied to verify the electrophysiological impact of the identified variants. A mouse model with cerebellar heterozygous knockout of the candidate gene was developed via adeno-associated virus injection, and dystonia-like phenotype inducement and rotarod tests were performed. In vivo multiunit electrical recording was applied to investigate the change in neural excitability in knockout mice. RESULTS Heterozygous variants of potassium inwardly rectifying channel subfamily J member 10 (KCNJ10) clustered in PKD patients were compared with those in the control groups. Fifteen variants were detected in 16 of 522 probands (frequency = 3.07%). Patients with KCNJ10 variants tended to have a milder manifestation compared to those with PRRT2 (proline-rich transmembrane protein 2) variants. KCNJ10 variants partially altered the transmembrane location of inwardly rectifying potassium channel 4.1 (Kir4.1). The Kcnj10 expression is consistent with the natural course of PKD. Variants resulted in different degrees of reduction in cell Kir4.1 currents, and mice with heterozygous conditional knockout of Kcnj10 in the cerebellum presented dystonic posture, together with poor motor coordination and motor learning ability in rotarod tests. The firing rate of deep cerebellar nuclei was significantly elevated in Kcnj10-cKO mice. CONCLUSION We identified heterozygous variants of KCNJ10 in PKD. Impaired function of Kir4.1 might lead to abnormal neuronal excitability, which attributed to PKD. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Xiaojun Huang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Xin Fu
- Center for Brain Science, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingying Wu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Xin Cheng
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Xiaoqi Hong
- Department of Obstetrics and Gynecology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyi Li
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Lan Zheng
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, China
| | - Qing Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shendi Chen
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Xiaorong Liu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xunhua Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoli Liu
- Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, China
| | - Zaiwei Zhou
- Shanghai Xunyin Biotechnology Co., Ltd., Shanghai, China
| | - Li Wu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kan Fang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Zhong
- Department of Neurology, Suzhou Hospital of Anhui Medical University, HeFei, China
| | - Mei Zhang
- Department of Neurology, The First Hospital Affiliated to Anhui University of Science and Technology, HeFei, China
| | - Xinghua Luan
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Wotu Tian
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
| | - Xiaoping Tong
- Department of Obstetrics and Gynecology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anatomy and Physiology, 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
- Shanghai Neurological Rare Disease Biobank and Precision Diagnostic Technical Service Platform, Shanghai, China
- Department of Neurology, Suzhou Hospital of Anhui Medical University, HeFei, China
- Department of Neurology, The First Hospital Affiliated to Anhui University of Science and Technology, HeFei, China
| |
Collapse
|
7
|
Salazar Leon LE, Kim LH, Sillitoe RV. Cerebellar deep brain stimulation as a dual-function therapeutic for restoring movement and sleep in dystonic mice. Neurotherapeutics 2024; 21:e00467. [PMID: 39448336 PMCID: PMC11585869 DOI: 10.1016/j.neurot.2024.e00467] [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: 11/11/2023] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
Dystonia arises with cerebellar dysfunction, which plays a key role in the emergence of multiple pathophysiological deficits that range from abnormal movements and postures to disrupted sleep. Current therapeutic interventions typically do not simultaneously address both the motor and non-motor symptoms of dystonia, underscoring the necessity for a multi-functional therapeutic strategy. Deep brain stimulation (DBS) is effectively used to reduce motor symptoms in dystonia, with existing parallel evidence arguing for its potential to correct sleep disturbances. However, the simultaneous efficacy of DBS for improving sleep and motor dysfunction, specifically by targeting the cerebellum, remains underexplored. Here, we test the effect of cerebellar DBS in two genetic mouse models with dystonia that exhibit sleep defects-Ptf1aCre;Vglut2fx/fx and Pdx1Cre;Vglut2fx/fx-which have overlapping cerebellar circuit miswiring defects but differing severity in motor phenotypes. By targeting DBS to the fiber tracts located between the cerebellar fastigial and the interposed nuclei (FN + INT-DBS), we modulated sleep dysfunction by enhancing sleep quality and timing. This DBS paradigm improved wakefulness and rapid eye movement sleep in both mutants. Additionally, the latency to reach REM sleep, a deficit observed in human dystonia patients, was reduced in both models. Cerebellar DBS also induced alterations in the electrocorticogram (ECoG) patterns that define sleep states. As expected, DBS reduced the severe dystonic twisting motor symptoms that are observed in the Ptf1aCre;Vglut2fx/fx mice. These findings highlight the potential for using cerebellar DBS to simultaneously improve sleep and reduce motor dysfunction in dystonia and uncover its potential as a dual-effect in vivo therapeutic strategy.
Collapse
Affiliation(s)
- Luis E Salazar Leon
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Linda H Kim
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Roy V Sillitoe
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Casoni F, Croci L, Marroni F, Demenego G, Marullo C, Cremona O, Codazzi F, Consalez GG. A spatial-temporal map of glutamatergic neurogenesis in the murine embryonic cerebellar nuclei uncovers a high degree of cellular heterogeneity. J Anat 2024; 245:560-571. [PMID: 38970393 PMCID: PMC11424815 DOI: 10.1111/joa.14107] [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: 12/18/2023] [Revised: 05/23/2024] [Accepted: 06/20/2024] [Indexed: 07/08/2024] Open
Abstract
The nuclei are the main output structures of the cerebellum. Each and every cerebellar cortical computation reaches several areas of the brain by means of cerebellar nuclei processing and integration. Nevertheless, our knowledge of these structures is still limited compared to the cerebellar cortex. Here, we present a mouse genetic inducible fate-mapping study characterizing rhombic lip-derived glutamatergic neurons of the nuclei, the most conspicuous family of long-range cerebellar efferent neurons. Glutamatergic neurons mainly occupy dorsal and lateral territories of the lateral and interposed nuclei, as well as the entire medial nucleus. In mice, they are born starting from about embryonic day 9.5, with a peak between 10.5 and 12.5, and invade the nuclei with a lateral-to-medial progression. While some markers label a heterogeneous population of neurons sharing a common location (BRN2), others appear to be lineage specific (TBR1, LMX1a, and MEIS2). A comparative analysis of TBR1 and LMX1a distributions reveals an incomplete overlap in their expression domains, in keeping with the existence of separate efferent subpopulations. Finally, some tagged glutamatergic progenitors are not labeled by any of the markers used in this study, disclosing further complexity. Taken together, our results obtained in late embryonic nuclei shed light on the heterogeneity of the excitatory neuron pool, underlying the diversity in connectivity and functions of this largely unexplored cerebellar territory. Our findings contribute to laying the groundwork for a comprehensive functional analysis of nuclear neuron subpopulations.
Collapse
Affiliation(s)
- Filippo Casoni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Laura Croci
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Marroni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Demenego
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Chiara Marullo
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ottavio Cremona
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Franca Codazzi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - G Giacomo Consalez
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
9
|
Li Z, Abram L, Peall KJ. Deciphering the Pathophysiological Mechanisms Underpinning Myoclonus Dystonia Using Pluripotent Stem Cell-Derived Cellular Models. Cells 2024; 13:1520. [PMID: 39329704 PMCID: PMC11430605 DOI: 10.3390/cells13181520] [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: 08/14/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 09/28/2024] Open
Abstract
Dystonia is a movement disorder with an estimated prevalence of 1.2% and is characterised by involuntary muscle contractions leading to abnormal postures and pain. Only symptomatic treatments are available with no disease-modifying or curative therapy, in large part due to the limited understanding of the underlying pathophysiology. However, the inherited monogenic forms of dystonia provide an opportunity for the development of disease models to examine these mechanisms. Myoclonus Dystonia, caused by SGCE mutations encoding the ε-sarcoglycan protein, represents one of now >50 monogenic forms. Previous research has implicated the involvement of the basal ganglia-cerebello-thalamo-cortical circuit in dystonia pathogenesis, but further work is needed to understand the specific molecular and cellular mechanisms. Pluripotent stem cell technology enables a patient-derived disease modelling platform harbouring disease-causing mutations. In this review, we discuss the current understanding of the aetiology of Myoclonus Dystonia, recent advances in producing distinct neuronal types from pluripotent stem cells, and their application in modelling Myoclonus Dystonia in vitro. Future research employing pluripotent stem cell-derived cellular models is crucial to elucidate how distinct neuronal types may contribute to dystonia and how disruption to neuronal function can give rise to dystonic disorders.
Collapse
Affiliation(s)
- Zongze Li
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK; (Z.L.); (L.A.)
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Laura Abram
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK; (Z.L.); (L.A.)
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Kathryn J. Peall
- Neuroscience and Mental Health Innovation Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK; (Z.L.); (L.A.)
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
10
|
Abstract
The cerebellum has a well-established role in controlling motor functions, including coordination, posture, and the learning of skilled movements. The mechanisms for how it carries out motor behavior remain under intense investigation. Interestingly though, in recent years the mechanisms of cerebellar function have faced additional scrutiny since nonmotor behaviors may also be controlled by the cerebellum. With such complexity arising, there is now a pressing need to better understand how cerebellar structure, function, and behavior intersect to influence behaviors that are dynamically called upon as an animal experiences its environment. Here, we discuss recent experimental work that frames possible neural mechanisms for how the cerebellum shapes disparate behaviors and why its dysfunction is catastrophic in hereditary and acquired conditions-both motor and nonmotor. For these reasons, the cerebellum might be the ideal therapeutic target.
Collapse
Affiliation(s)
- Linda H Kim
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA;
| | - Detlef H Heck
- Center for Cerebellar Network Structure and Function in Health and Disease, University of Minnesota, Duluth, Minnesota, USA
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota, USA
| | - Roy V Sillitoe
- Departments of Neuroscience and Pediatrics, Program in Developmental Biology, and Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA;
| |
Collapse
|
11
|
Gittis AH, Sillitoe RV. Circuit-Specific Deep Brain Stimulation Provides Insights into Movement Control. Annu Rev Neurosci 2024; 47:63-83. [PMID: 38424473 DOI: 10.1146/annurev-neuro-092823-104810] [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] [Indexed: 03/02/2024]
Abstract
Deep brain stimulation (DBS), a method in which electrical stimulation is delivered to specific areas of the brain, is an effective treatment for managing symptoms of a number of neurological and neuropsychiatric disorders. Clinical access to neural circuits during DBS provides an opportunity to study the functional link between neural circuits and behavior. This review discusses how the use of DBS in Parkinson's disease and dystonia has provided insights into the brain networks and physiological mechanisms that underlie motor control. In parallel, insights from basic science about how patterns of electrical stimulation impact plasticity and communication within neural circuits are transforming DBS from a therapy for treating symptoms to a therapy for treating circuits, with the goal of training the brain out of its diseased state.
Collapse
Affiliation(s)
- Aryn H Gittis
- Department of Biological Sciences and Neuroscience Institute, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA;
| | - Roy V Sillitoe
- Departments of Neuroscience, Pathology & Immunology, and Pediatrics; and Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
12
|
van der Heijden ME, Brown AM, Kizek DJ, Sillitoe RV. Cerebellar nuclei cells produce distinct pathogenic spike signatures in mouse models of ataxia, dystonia, and tremor. eLife 2024; 12:RP91483. [PMID: 39072369 DOI: 10.7554/elife.91483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
The cerebellum contributes to a diverse array of motor conditions, including ataxia, dystonia, and tremor. The neural substrates that encode this diversity are unclear. Here, we tested whether the neural spike activity of cerebellar output neurons is distinct between movement disorders with different impairments, generalizable across movement disorders with similar impairments, and capable of causing distinct movement impairments. Using in vivo awake recordings as input data, we trained a supervised classifier model to differentiate the spike parameters between mouse models for ataxia, dystonia, and tremor. The classifier model correctly assigned mouse phenotypes based on single-neuron signatures. Spike signatures were shared across etiologically distinct but phenotypically similar disease models. Mimicking these pathophysiological spike signatures with optogenetics induced the predicted motor impairments in otherwise healthy mice. These data show that distinct spike signatures promote the behavioral presentation of cerebellar diseases.
Collapse
Affiliation(s)
- Meike E van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Amanda M Brown
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Dominic J Kizek
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Roy V Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
- Department of Pediatrics, Baylor College of Medicine, Houston, United States
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| |
Collapse
|
13
|
Yoshioka N, Kurose M, Sano H, Tran DM, Chiken S, Tainaka K, Yamamura K, Kobayashi K, Nambu A, Takebayashi H. Sensory-motor circuit is a therapeutic target for dystonia musculorum mice, a model of hereditary sensory and autonomic neuropathy 6. SCIENCE ADVANCES 2024; 10:eadj9335. [PMID: 39058787 PMCID: PMC11277474 DOI: 10.1126/sciadv.adj9335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 06/25/2024] [Indexed: 07/28/2024]
Abstract
Mutations in Dystonin (DST), which encodes cytoskeletal linker proteins, cause hereditary sensory and autonomic neuropathy 6 (HSAN-VI) in humans and the dystonia musculorum (dt) phenotype in mice; however, the neuronal circuit underlying the HSAN-VI and dt phenotype is unresolved. dt mice exhibit dystonic movements accompanied by the simultaneous contraction of agonist and antagonist muscles and postnatal lethality. Here, we identified the sensory-motor circuit as a major causative neural circuit using a gene trap system that enables neural circuit-selective inactivation and restoration of Dst by Cre-mediated recombination. Sensory neuron-selective Dst deletion led to motor impairment, degeneration of proprioceptive sensory neurons, and disruption of the sensory-motor circuit. Restoration of Dst expression in sensory neurons using Cre driver mice or a single postnatal injection of Cre-expressing adeno-associated virus ameliorated sensory degeneration and improved abnormal movements. These findings demonstrate that the sensory-motor circuit is involved in the movement disorders in dt mice and that the sensory circuit is a therapeutic target for HSAN-VI.
Collapse
Affiliation(s)
- Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Transdisciplinary Research Programs, Niigata University, Niigata, Japan
| | - Masayuki Kurose
- Department of Physiology, School of Dentistry, Iwate Medical University, Yahaba, Japan
- Division of Oral Physiology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiromi Sano
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences, SOKENDAI, Okazaki, Japan
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Dang Minh Tran
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Satomi Chiken
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences, SOKENDAI, Okazaki, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kensuke Yamamura
- Division of Oral Physiology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Japan
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences, SOKENDAI, Okazaki, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Center for Coordination of Research Facilities, Niigata University, Niigata, Japan
| |
Collapse
|
14
|
Salazar Leon LE, Brown AM, Kaku H, Sillitoe RV. Purkinje cell dysfunction causes disrupted sleep in ataxic mice. Dis Model Mech 2024; 17:dmm050379. [PMID: 38563553 PMCID: PMC11190574 DOI: 10.1242/dmm.050379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Purkinje cell dysfunction disrupts movement and causes disorders such as ataxia. Recent evidence suggests that Purkinje cell dysfunction may also alter sleep regulation. Here, we used an ataxic mouse model generated by silencing Purkinje cell neurotransmission (L7Cre;Vgatfx/fx) to better understand how cerebellar dysfunction impacts sleep physiology. We focused our analysis on sleep architecture and electrocorticography (ECoG) patterns based on their relevance to extracting physiological measurements during sleep. We found that circadian activity was unaltered in the mutant mice, although their sleep parameters and ECoG patterns were modified. The L7Cre;Vgatfx/fx mutant mice had decreased wakefulness and rapid eye movement (REM) sleep, whereas non-REM sleep was increased. The mutants had an extended latency to REM sleep, which is also observed in human patients with ataxia. Spectral analysis of ECoG signals revealed alterations in the power distribution across different frequency bands defining sleep. Therefore, Purkinje cell dysfunction may influence wakefulness and equilibrium of distinct sleep stages in ataxia. Our findings posit a connection between cerebellar dysfunction and disrupted sleep and underscore the importance of examining cerebellar circuit function in sleep disorders.
Collapse
Affiliation(s)
- Luis E. Salazar Leon
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Amanda M. Brown
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Heet Kaku
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Roy V. Sillitoe
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
15
|
Nguyen MX, Brown AM, Lin T, Sillitoe RV, Gill JS. Targeting DBS to the centrolateral thalamic nucleus improves movement in a lesion-based model of acquired cerebellar dystonia in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595095. [PMID: 38826430 PMCID: PMC11142135 DOI: 10.1101/2024.05.21.595095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Dystonia is the third most common movement disorder and an incapacitating co-morbidity in a variety of neurologic conditions. Dystonia can be caused by genetic, degenerative, idiopathic, and acquired etiologies, which are hypothesized to converge on a "dystonia network" consisting of the basal ganglia, thalamus, cerebellum, and cerebral cortex. In acquired dystonia, focal lesions to subcortical areas in the network - the basal ganglia, thalamus, and cerebellum - lead to a dystonia that can be difficult to manage with canonical treatments, including deep brain stimulation (DBS). While studies in animal models have begun to parse the contribution of individual nodes in the dystonia network, how acquired injury to the cerebellar outflow tracts instigates dystonia; and how network modulation interacts with symptom latency remain as unexplored questions. Here, we present an electrolytic lesioning paradigm that bilaterally targets the cerebellar outflow tracts. We found that lesioning these tracts, at the junction of the superior cerebellar peduncles and the medial and intermediate cerebellar nuclei, resulted in acute, severe dystonia. We observed that dystonia is reduced with one hour of DBS of the centrolateral thalamic nucleus, a first order node in the network downstream of the cerebellar nuclei. In contrast, one hour of stimulation at a second order node in the short latency, disynaptic projection from the cerebellar nuclei, the striatum, did not modulate the dystonia in the short-term. Our study introduces a robust paradigm for inducing acute, severe dystonia, and demonstrates that targeted modulation based on network principles powerfully rescues motor behavior. These data inspire the identification of therapeutic targets for difficult to manage acquired dystonia.
Collapse
Affiliation(s)
- Megan X. Nguyen
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| | - Amanda M. Brown
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Tao Lin
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Roy V. Sillitoe
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Jason S. Gill
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
16
|
Matsuda T, Morigaki R, Hayasawa H, Koyama H, Oda T, Miyake K, Takagi Y. Striatal parvalbumin interneurons are activated in a mouse model of cerebellar dystonia. Dis Model Mech 2024; 17:dmm050338. [PMID: 38616770 PMCID: PMC11128288 DOI: 10.1242/dmm.050338] [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: 06/03/2023] [Accepted: 04/09/2024] [Indexed: 04/16/2024] Open
Abstract
Dystonia is thought to arise from abnormalities in the motor loop of the basal ganglia; however, there is an ongoing debate regarding cerebellar involvement. We adopted an established cerebellar dystonia mouse model by injecting ouabain to examine the contribution of the cerebellum. Initially, we examined whether the entopeduncular nucleus (EPN), substantia nigra pars reticulata (SNr), globus pallidus externus (GPe) and striatal neurons were activated in the model. Next, we examined whether administration of a dopamine D1 receptor agonist and dopamine D2 receptor antagonist or selective ablation of striatal parvalbumin (PV, encoded by Pvalb)-expressing interneurons could modulate the involuntary movements of the mice. The cerebellar dystonia mice had a higher number of cells positive for c-fos (encoded by Fos) in the EPN, SNr and GPe, as well as a higher positive ratio of c-fos in striatal PV interneurons, than those in control mice. Furthermore, systemic administration of combined D1 receptor agonist and D2 receptor antagonist and selective ablation of striatal PV interneurons relieved the involuntary movements of the mice. Abnormalities in the motor loop of the basal ganglia could be crucially involved in cerebellar dystonia, and modulating PV interneurons might provide a novel treatment strategy.
Collapse
Affiliation(s)
- Taku Matsuda
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Ryoma Morigaki
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
- Department of Advanced Brain Research, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
- Parkinson's Disease and Dystonia Research Center, Tokushima University Hospital, Tokushima 770-8503, Japan
| | - Hiroaki Hayasawa
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Hiroshi Koyama
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Teruo Oda
- Department of Advanced Brain Research, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Kazuhisa Miyake
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Yasushi Takagi
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
- Department of Advanced Brain Research, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| |
Collapse
|
17
|
van der Heijden ME, Brown AM, Kizek DJ, Sillitoe RV. Cerebellar nuclei cells produce distinct pathogenic spike signatures in mouse models of ataxia, dystonia, and tremor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.07.539767. [PMID: 37214855 PMCID: PMC10197583 DOI: 10.1101/2023.05.07.539767] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The cerebellum contributes to a diverse array of motor conditions including ataxia, dystonia, and tremor. The neural substrates that encode this diversity are unclear. Here, we tested whether the neural spike activity of cerebellar output neurons is distinct between movement disorders with different impairments, generalizable across movement disorders with similar impairments, and capable of causing distinct movement impairments. Using in vivo awake recordings as input data, we trained a supervised classifier model to differentiate the spike parameters between mouse models for ataxia, dystonia, and tremor. The classifier model correctly assigned mouse phenotypes based on single neuron signatures. Spike signatures were shared across etiologically distinct but phenotypically similar disease models. Mimicking these pathophysiological spike signatures with optogenetics induced the predicted motor impairments in otherwise healthy mice. These data show that distinct spike signatures promote the behavioral presentation of cerebellar diseases.
Collapse
|
18
|
Kebschull JM, Casoni F, Consalez GG, Goldowitz D, Hawkes R, Ruigrok TJH, Schilling K, Wingate R, Wu J, Yeung J, Uusisaari MY. Cerebellum Lecture: the Cerebellar Nuclei-Core of the Cerebellum. CEREBELLUM (LONDON, ENGLAND) 2024; 23:620-677. [PMID: 36781689 PMCID: PMC10951048 DOI: 10.1007/s12311-022-01506-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/10/2022] [Indexed: 02/15/2023]
Abstract
The cerebellum is a key player in many brain functions and a major topic of neuroscience research. However, the cerebellar nuclei (CN), the main output structures of the cerebellum, are often overlooked. This neglect is because research on the cerebellum typically focuses on the cortex and tends to treat the CN as relatively simple output nuclei conveying an inverted signal from the cerebellar cortex to the rest of the brain. In this review, by adopting a nucleocentric perspective we aim to rectify this impression. First, we describe CN anatomy and modularity and comprehensively integrate CN architecture with its highly organized but complex afferent and efferent connectivity. This is followed by a novel classification of the specific neuronal classes the CN comprise and speculate on the implications of CN structure and physiology for our understanding of adult cerebellar function. Based on this thorough review of the adult literature we provide a comprehensive overview of CN embryonic development and, by comparing cerebellar structures in various chordate clades, propose an interpretation of CN evolution. Despite their critical importance in cerebellar function, from a clinical perspective intriguingly few, if any, neurological disorders appear to primarily affect the CN. To highlight this curious anomaly, and encourage future nucleocentric interpretations, we build on our review to provide a brief overview of the various syndromes in which the CN are currently implicated. Finally, we summarize the specific perspectives that a nucleocentric view of the cerebellum brings, move major outstanding issues in CN biology to the limelight, and provide a roadmap to the key questions that need to be answered in order to create a comprehensive integrated model of CN structure, function, development, and evolution.
Collapse
Affiliation(s)
- Justus M Kebschull
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | - Filippo Casoni
- Division of Neuroscience, San Raffaele Scientific Institute, and San Raffaele University, Milan, Italy
| | - G Giacomo Consalez
- Division of Neuroscience, San Raffaele Scientific Institute, and San Raffaele University, Milan, Italy
| | - Daniel Goldowitz
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Richard Hawkes
- Department of Cell Biology & Anatomy and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Tom J H Ruigrok
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Karl Schilling
- Department of Anatomy, Anatomy & Cell Biology, Rheinische Friedrich-Wilhelms-Universität, 53115, Bonn, Federal Republic of Germany
| | - Richard Wingate
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Joshua Wu
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Joanna Yeung
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Marylka Yoe Uusisaari
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-Son, Kunigami-Gun, Okinawa, 904-0495, Japan.
| |
Collapse
|
19
|
Kumar G, Zhou Z, Wang Z, Kwan KM, Tin C, Ma CHE. Real-time field-programmable gate array-based closed-loop deep brain stimulation platform targeting cerebellar circuitry rescues motor deficits in a mouse model of cerebellar ataxia. CNS Neurosci Ther 2024; 30:e14638. [PMID: 38488445 PMCID: PMC10941591 DOI: 10.1111/cns.14638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 01/09/2024] [Accepted: 02/01/2024] [Indexed: 03/18/2024] Open
Abstract
AIMS The open-loop nature of conventional deep brain stimulation (DBS) produces continuous and excessive stimulation to patients which contributes largely to increased prevalence of adverse side effects. Cerebellar ataxia is characterized by abnormal Purkinje cells (PCs) dendritic arborization, loss of PCs and motor coordination, and muscle weakness with no effective treatment. We aim to develop a real-time field-programmable gate array (FPGA) prototype targeting the deep cerebellar nuclei (DCN) to close the loop for ataxia using conditional double knockout mice with deletion of PC-specific LIM homeobox (Lhx)1 and Lhx5, resulting in abnormal dendritic arborization and motor deficits. METHODS We implanted multielectrode array in the DCN and muscles of ataxia mice. The beneficial effect of open-loop DCN-DBS or closed-loop DCN-DBS was compared by motor behavioral assessments, electromyography (EMG), and neural activities (neurospike and electroencephalogram) in freely moving mice. FPGA board, which performed complex real-time computation, was used for closed-loop DCN-DBS system. RESULTS Closed-loop DCN-DBS was triggered only when symptomatic muscle EMG was detected in a real-time manner, which restored motor activities, electroencephalogram activities and neurospike properties completely in ataxia mice. Closed-loop DCN-DBS was more effective than an open-loop paradigm as it reduced the frequency of DBS. CONCLUSION Our real-time FPGA-based DCN-DBS system could be a potential clinical strategy for alleviating cerebellar ataxia and other movement disorders.
Collapse
Affiliation(s)
- Gajendra Kumar
- Department of NeuroscienceCity University of Hong KongHong KongHong Kong SAR
| | - Zhanhong Zhou
- Department of Biomedical EngineeringCity University of Hong KongHong KongHong Kong SAR
| | - Zhihua Wang
- Department of Biomedical EngineeringCity University of Hong KongHong KongHong Kong SAR
| | - Kin Ming Kwan
- School of Life Sciences, Center for Cell and Developmental Biology and State Key Laboratory of AgrobiotechnologyThe Chinese University of Hong KongHong KongHong Kong SAR
| | - Chung Tin
- Department of Biomedical EngineeringCity University of Hong KongHong KongHong Kong SAR
| | - Chi Him Eddie Ma
- Department of NeuroscienceCity University of Hong KongHong KongHong Kong SAR
| |
Collapse
|
20
|
Abstract
Dystonia is a clinically and genetically highly heterogeneous neurological disorder characterized by abnormal movements and postures caused by involuntary sustained or intermittent muscle contractions. A number of groundbreaking genetic and molecular insights have recently been gained. While they enable genetic testing and counseling, their translation into new therapies is still limited. However, we are beginning to understand shared pathophysiological pathways and molecular mechanisms. It has become clear that dystonia results from a dysfunctional network involving the basal ganglia, cerebellum, thalamus, and cortex. On the molecular level, more than a handful of, often intertwined, pathways have been linked to pathogenic variants in dystonia genes, including gene transcription during neurodevelopment (e.g., KMT2B, THAP1), calcium homeostasis (e.g., ANO3, HPCA), striatal dopamine signaling (e.g., GNAL), endoplasmic reticulum stress response (e.g., EIF2AK2, PRKRA, TOR1A), autophagy (e.g., VPS16), and others. Thus, different forms of dystonia can be molecularly grouped, which may facilitate treatment development in the future.
Collapse
Affiliation(s)
- Mirja Thomsen
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Lara M Lange
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| |
Collapse
|
21
|
Zhai P, Romano V, Soggia G, Bauer S, van Wingerden N, Jacobs T, van der Horst A, White JJ, Mazza R, De Zeeuw CI. Whisker kinematics in the cerebellum. J Physiol 2024; 602:153-181. [PMID: 37987552 DOI: 10.1113/jp284064] [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: 11/07/2022] [Accepted: 10/30/2023] [Indexed: 11/22/2023] Open
Abstract
The whisker system is widely used as a model system for understanding sensorimotor integration. Purkinje cells in the crus regions of the cerebellum have been reported to linearly encode whisker midpoint, but it is unknown whether the paramedian and simplex lobules as well as their target neurons in the cerebellar nuclei also encode whisker kinematics and if so which ones. Elucidating how these kinematics are represented throughout the cerebellar hemisphere is essential for understanding how the cerebellum coordinates multiple sensorimotor modalities. Exploring the cerebellar hemisphere of mice using optogenetic stimulation, we found that whisker movements can be elicited by stimulation of Purkinje cells in not only crus1 and crus2, but also in the paramedian lobule and lobule simplex; activation of cells in the medial paramedian lobule had on average the shortest latency, whereas that of cells in lobule simplex elicited similar kinematics as those in crus1 and crus2. During spontaneous whisking behaviour, simple spike activity correlated in general better with velocity than position of the whiskers, but it varied between protraction and retraction as well as per lobule. The cerebellar nuclei neurons targeted by the Purkinje cells showed similar activity patterns characterized by a wide variety of kinematic signals, yet with a dominance for velocity. Taken together, our data indicate that whisker movements are much more prominently and diversely represented in the cerebellar cortex and nuclei than assumed, highlighting the rich repertoire of cerebellar control in the kinematics of movements that can be engaged during coordination. KEY POINTS: Excitation of Purkinje cells throughout the cerebellar hemispheres induces whisker movement, with the shortest latency and longest duration within the paramedian lobe. Purkinje cells have differential encoding for the fast and slow components of whisking. Purkinje cells encode not only the position but also the velocity of whiskers. Purkinje cells with high sensitivity for whisker velocity are preferentially located in the medial part of lobule simplex, crus1 and lateral paramedian. In the downstream cerebellar nuclei, neurons with high sensitivity for whisker velocity are located at the intersection between the medial and interposed nucleus.
Collapse
Affiliation(s)
- Peipei Zhai
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Vincenzo Romano
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Giulia Soggia
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Staf Bauer
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | - Thomas Jacobs
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | - Joshua J White
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Roberta Mazza
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- Netherlands Institute for Neuroscience, Royal Dutch Academy of Arts & Sciences, Amsterdam, Netherlands
| |
Collapse
|
22
|
Gill JS, Nguyen MX, Hull M, van der Heijden ME, Nguyen K, Thomas SP, Sillitoe RV. Function and dysfunction of the dystonia network: an exploration of neural circuits that underlie the acquired and isolated dystonias. DYSTONIA 2023; 2:11805. [PMID: 38273865 PMCID: PMC10810232 DOI: 10.3389/dyst.2023.11805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Dystonia is a highly prevalent movement disorder that can manifest at any time across the lifespan. An increasing number of investigations have tied this disorder to dysfunction of a broad "dystonia network" encompassing the cerebellum, thalamus, basal ganglia, and cortex. However, pinpointing how dysfunction of the various anatomic components of the network produces the wide variety of dystonia presentations across etiologies remains a difficult problem. In this review, a discussion of functional network findings in non-mendelian etiologies of dystonia is undertaken. Initially acquired etiologies of dystonia and how lesion location leads to alterations in network function are explored, first through an examination of cerebral palsy, in which early brain injury may lead to dystonic/dyskinetic forms of the movement disorder. The discussion of acquired etiologies then continues with an evaluation of the literature covering dystonia resulting from focal lesions followed by the isolated focal dystonias, both idiopathic and task dependent. Next, how the dystonia network responds to therapeutic interventions, from the "geste antagoniste" or "sensory trick" to botulinum toxin and deep brain stimulation, is covered with an eye towards finding similarities in network responses with effective treatment. Finally, an examination of how focal network disruptions in mouse models has informed our understanding of the circuits involved in dystonia is provided. Together, this article aims to offer a synthesis of the literature examining dystonia from the perspective of brain networks and it provides grounding for the perspective of dystonia as disorder of network function.
Collapse
Affiliation(s)
- Jason S. Gill
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
| | - Megan X. Nguyen
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
| | - Mariam Hull
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Meike E. van der Heijden
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United State
| | - Ken Nguyen
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United State
| | - Sruthi P. Thomas
- H. Ben Taub Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, United States
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
| | - Roy V. Sillitoe
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United State
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
23
|
van der Heijden ME, Sillitoe RV. Cerebellar dysfunction in rodent models with dystonia, tremor, and ataxia. DYSTONIA 2023; 2:11515. [PMID: 38105800 PMCID: PMC10722573 DOI: 10.3389/dyst.2023.11515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Dystonia is a movement disorder characterized by involuntary co- or over-contractions of the muscles, which results in abnormal postures and movements. These symptoms arise from the pathophysiology of a brain-wide dystonia network. There is mounting evidence suggesting that the cerebellum is a central node in this network. For example, manipulations that target the cerebellum cause dystonic symptoms in mice, and cerebellar neuromodulation reduces these symptoms. Although numerous findings provide insight into dystonia pathophysiology, they also raise further questions. Namely, how does cerebellar pathophysiology cause the diverse motor abnormalities in dystonia, tremor, and ataxia? Here, we describe recent work in rodents showing that distinct cerebellar circuit abnormalities could define different disorders and we discuss potential mechanisms that determine the behavioral presentation of cerebellar diseases.
Collapse
Affiliation(s)
- 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 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
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, United States
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
24
|
Leon LES, Kim LH, Sillitoe RV. Cerebellar deep brain stimulation as a dual-function therapeutic for restoring movement and sleep in dystonic mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564790. [PMID: 37961355 PMCID: PMC10635001 DOI: 10.1101/2023.10.30.564790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dystonia arises with cerebellar dysfunction, which plays a key role in the emergence of multiple pathophysiological deficits that range from abnormal movements and postures to disrupted sleep. Current therapeutic interventions typically do not simultaneously address both the motor and non-motor (sleep-related) symptoms of dystonia, underscoring the necessity for a multi-functional therapeutic strategy. Deep brain stimulation (DBS) is effectively used to reduce motor symptoms in dystonia, with existing parallel evidence arguing for its potential to correct sleep disturbances. However, the simultaneous efficacy of DBS for improving sleep and motor dysfunction, specifically by targeting the cerebellum, remains underexplored. Here, we test the effect of cerebellar DBS in two genetic mouse models with dystonia that exhibit sleep defects- Ptf1a Cre ;Vglut2 fx/fx and Pdx1 Cre ;Vglut2 fx/fx -which have overlapping cerebellar circuit miswiring defects but differing severity in motor phenotypes. By targeting DBS to the cerebellar fastigial and interposed nuclei, we modulated sleep dysfunction by enhancing sleep quality and timing in both models. This DBS paradigm improved wakefulness (decreased) and rapid eye movement (REM) sleep (increased) in both mutants. Additionally, the latency to reach REM sleep, a deficit observed in human dystonia patients, was reduced in both models. Cerebellar DBS also induced alterations in the electrocorticogram (ECoG) patterns that define sleep states. As expected, DBS reduced the severe dystonic twisting motor symptoms that are observed in the Ptf1a Cre ;Vglut2 fx/fx mutant mice. These findings highlight the potential for using cerebellar DBS to improve sleep and reduce motor dysfunction in dystonia and uncover its potential as a dual-effect in vivo therapeutic strategy.
Collapse
|
25
|
Brown AM, van der Heijden ME, Jinnah HA, Sillitoe RV. Cerebellar Dysfunction as a Source of Dystonic Phenotypes in Mice. CEREBELLUM (LONDON, ENGLAND) 2023; 22:719-729. [PMID: 35821365 PMCID: PMC10307717 DOI: 10.1007/s12311-022-01441-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
There is now a substantial amount of compelling evidence demonstrating that the cerebellum may be a central locus in dystonia pathogenesis. Studies using spontaneous genetic mutations in rats and mice, engineered genetic alleles in mice, shRNA knockdown in mice, and conditional genetic silencing of fast neurotransmission in mice have all uncovered a common set of behavioral and electrophysiological defects that point to cerebellar cortical and cerebellar nuclei dysfunction as a source of dystonic phenotypes. Here, we revisit the Ptf1aCre/+;Vglut2flox/flox mutant mouse to define fundamental phenotypes and measures that are valuable for testing the cellular, circuit, and behavioral mechanisms that drive dystonia. In this model, excitatory neurotransmission from climbing fibers is genetically eliminated and, as a consequence, Purkinje cell and cerebellar nuclei firing are altered in vivo, with a prominent and lasting irregular burst pattern of spike activity in cerebellar nuclei neurons. The resulting impact on behavior is that the mice have developmental abnormalities, including twisting of the limbs and torso. These behaviors continue into adulthood along with a tremor, which can be measured with a tremor monitor or EMG. Importantly, expression of dystonic behavior is reduced upon cerebellar-targeted deep brain stimulation. The presence of specific combinations of disease-like features and therapeutic responses could reveal the causative mechanisms of different types of dystonia and related conditions. Ultimately, an emerging theme places cerebellar dysfunction at the center of a broader dystonia brain network.
Collapse
Affiliation(s)
- Amanda M Brown
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - Meike E van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA
| | - H A Jinnah
- Departments of Neurology, Human Genetics and Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Roy V Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, 77030, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
26
|
Beckinghausen J, Ortiz-Guzman J, Lin T, Bachman B, Salazar Leon LE, Liu Y, Heck DH, Arenkiel BR, Sillitoe RV. The cerebellum contributes to generalized seizures by altering activity in the ventral posteromedial nucleus. Commun Biol 2023; 6:731. [PMID: 37454228 PMCID: PMC10349834 DOI: 10.1038/s42003-023-05100-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/05/2023] [Indexed: 07/18/2023] Open
Abstract
Thalamo-cortical networks are central to seizures, yet it is unclear how these circuits initiate seizures. We test whether a facial region of the thalamus, the ventral posteromedial nucleus (VPM), is a source of generalized, convulsive motor seizures and if convergent VPM input drives the behavior. To address this question, we devise an in vivo optogenetic mouse model to elicit convulsive motor seizures by driving these inputs and perform single-unit recordings during awake, convulsive seizures to define the local activity of thalamic neurons before, during, and after seizure onset. We find dynamic activity with biphasic properties, raising the possibility that heterogenous activity promotes seizures. Virus tracing identifies cerebellar and cerebral cortical afferents as robust contributors to the seizures. Of these inputs, only microinfusion of lidocaine into the cerebellar nuclei blocks seizure initiation. Our data reveal the VPM as a source of generalized convulsive seizures, with cerebellar input providing critical signals.
Collapse
Affiliation(s)
- Jaclyn Beckinghausen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
| | - Joshua Ortiz-Guzman
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - Tao Lin
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
| | - Benjamin Bachman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Luis E Salazar Leon
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
| | - Yu Liu
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, 103515 University Dr., Duluth, MN, USA
| | - Detlef H Heck
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, 103515 University Dr., Duluth, MN, USA
| | - Benjamin R Arenkiel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, USA.
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
27
|
Tsagkaris S, Yau EKC, McClelland V, Papandreou A, Siddiqui A, Lumsden DE, Kaminska M, Guedj E, Hammers A, Lin JP. Metabolic patterns in brain 18F-fluorodeoxyglucose PET relate to aetiology in paediatric dystonia. Brain 2023; 146:2512-2523. [PMID: 36445406 PMCID: PMC10232264 DOI: 10.1093/brain/awac439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/24/2022] [Accepted: 11/08/2022] [Indexed: 12/09/2023] Open
Abstract
There is a lack of imaging markers revealing the functional characteristics of different brain regions in paediatric dystonia. In this observational study, we assessed the utility of [18F]2-fluoro-2-deoxy-D-glucose (FDG)-PET in understanding dystonia pathophysiology by revealing specific resting awake brain glucose metabolism patterns in different childhood dystonia subgroups. PET scans from 267 children with dystonia being evaluated for possible deep brain stimulation surgery between September 2007 and February 2018 at Evelina London Children's Hospital (ELCH), UK, were examined. Scans without gross anatomical abnormality (e.g. large cysts, significant ventriculomegaly; n = 240) were analysed with Statistical Parametric Mapping (SPM12). Glucose metabolism patterns were examined in the 144/240 (60%) cases with the 10 commonest childhood-onset dystonias, focusing on nine anatomical regions. A group of 39 adult controls was used for comparisons. The genetic dystonias were associated with the following genes: TOR1A, THAP1, SGCE, KMT2B, HPRT1 (Lesch Nyhan disease), PANK2 and GCDH (Glutaric Aciduria type 1). The acquired cerebral palsy (CP) cases were divided into those related to prematurity (CP-Preterm), neonatal jaundice/kernicterus (CP-Kernicterus) and hypoxic-ischaemic encephalopathy (CP-Term). Each dystonia subgroup had distinct patterns of altered FDG-PET uptake. Focal glucose hypometabolism of the pallidi, putamina or both, was the commonest finding, except in PANK2, where basal ganglia metabolism appeared normal. HPRT1 uniquely showed glucose hypometabolism across all nine cerebral regions. Temporal lobe glucose hypometabolism was found in KMT2B, HPRT1 and CP-Kernicterus. Frontal lobe hypometabolism was found in SGCE, HPRT1 and PANK2. Thalamic and brainstem hypometabolism were seen only in HPRT1, CP-Preterm and CP-term dystonia cases. The combination of frontal and parietal lobe hypermetabolism was uniquely found in CP-term cases. PANK2 cases showed a distinct combination of parietal hypermetabolism with cerebellar hypometabolism but intact putaminal-pallidal glucose metabolism. HPRT1, PANK2, CP-kernicterus and CP-preterm cases had cerebellar and insula glucose hypometabolism as well as parietal glucose hypermetabolism. The study findings offer insights into the pathophysiology of dystonia and support the network theory for dystonia pathogenesis. 'Signature' patterns for each dystonia subgroup could be a useful biomarker to guide differential diagnosis and inform personalized management strategies.
Collapse
Affiliation(s)
- Stavros Tsagkaris
- Children’s Neurosciences, Complex Motor Disorders Service (CMDS), Evelina London Children's Hospital, Guy's and St Thomas’ NHS Foundation Trust (GSTT), London SE1 7EH, UK
- King’s College London & Guy’s and St Thomas’ PET Centre, Division of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, UK
| | - Eric K C Yau
- Department of Paediatrics & Adolescent Medicine, Princess Margaret Hospital, Kowloon, Hong Kong
| | - Verity McClelland
- Children’s Neurosciences, Complex Motor Disorders Service (CMDS), Evelina London Children's Hospital, Guy's and St Thomas’ NHS Foundation Trust (GSTT), London SE1 7EH, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Apostolos Papandreou
- Children’s Neurosciences, Complex Motor Disorders Service (CMDS), Evelina London Children's Hospital, Guy's and St Thomas’ NHS Foundation Trust (GSTT), London SE1 7EH, UK
- Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, University College London Great Ormond Street Institute of Child Health, London WC1N 1DZ, UK
| | - Ata Siddiqui
- Neuroradiology Department, Evelina London Children's Hospital, Guy's and St Thomas’ NHS Foundation Trust (GSTT), London SE1 7EH, UK
| | - Daniel E Lumsden
- Children’s Neurosciences, Complex Motor Disorders Service (CMDS), Evelina London Children's Hospital, Guy's and St Thomas’ NHS Foundation Trust (GSTT), London SE1 7EH, UK
- Perinatal Imaging, Division of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, UK
| | - Margaret Kaminska
- Children’s Neurosciences, Complex Motor Disorders Service (CMDS), Evelina London Children's Hospital, Guy's and St Thomas’ NHS Foundation Trust (GSTT), London SE1 7EH, UK
| | - Eric Guedj
- CERIMED, Nuclear Medicine Department, Aix Marseille Universite, APHM, CNRS, Centrale Marseille, Institut Fresnel, Timone Hospital, 13397 Marseille, France
| | - Alexander Hammers
- King’s College London & Guy’s and St Thomas’ PET Centre, Division of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, UK
| | - Jean-Pierre Lin
- Children’s Neurosciences, Complex Motor Disorders Service (CMDS), Evelina London Children's Hospital, Guy's and St Thomas’ NHS Foundation Trust (GSTT), London SE1 7EH, UK
- Women and Children’s Health Institute Faculty of Life Sciences & Medicine, Kings Health Partners, King’s College London, London SE1 7EH, UK
| |
Collapse
|
28
|
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
|
29
|
van der Heijden ME, Rey Hipolito AG, Kim LH, Kizek DJ, Perez RM, Lin T, Sillitoe RV. Glutamatergic cerebellar neurons differentially contribute to the acquisition of motor and social behaviors. Nat Commun 2023; 14:2771. [PMID: 37188723 PMCID: PMC10185563 DOI: 10.1038/s41467-023-38475-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
Insults to the developing cerebellum can cause motor, language, and social deficits. Here, we investigate whether developmental insults to different cerebellar neurons constrain the ability to acquire cerebellar-dependent behaviors. We perturb cerebellar cortical or nuclei neuron function by eliminating glutamatergic neurotransmission during development, and then we measure motor and social behaviors in early postnatal and adult mice. Altering cortical and nuclei neurons impacts postnatal motor control and social vocalizations. Normalizing neurotransmission in cortical neurons but not nuclei neurons restores social behaviors while the motor deficits remain impaired in adults. In contrast, manipulating only a subset of nuclei neurons leaves social behaviors intact but leads to early motor deficits that are restored by adulthood. Our data uncover that glutamatergic neurotransmission from cerebellar cortical and nuclei neurons differentially control the acquisition of motor and social behaviors, and that the brain can compensate for some but not all perturbations to the developing cerebellum.
Collapse
Affiliation(s)
- Meike E van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Alejandro G Rey Hipolito
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Linda H Kim
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Dominic J Kizek
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Ross M Perez
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Tao Lin
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Roy V Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
30
|
Lycett M, Cui CK, Dragicevich D, Harris R, Ng K. Hypertrophic olivary degeneration associated with bilateral vocal cord adductor dystonia. BMC Neurol 2023; 23:105. [PMID: 36918827 PMCID: PMC10012681 DOI: 10.1186/s12883-023-03123-8] [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: 08/30/2022] [Accepted: 02/15/2023] [Indexed: 03/15/2023] Open
Abstract
BACKGROUND Hypertrophic olivary degeneration (HOD) is a rare condition caused by lesions within the dentato-rubro-olivary pathway, resulting in ocular nystagmus and palatal myoclonus (oculopalatal tremor) but not usually dystonia. Dystonia is an uncommon association, and we present the first reported association of hypertrophic olivary degeneration with bilateral vocal cord dystonia. CASE PRESENTATION A 33 year old male presented initially with acute hydrocephalus on the background of previous ventriculoperitoneal (VP) shunting for previously treated medulloblastoma. After revision of the VP shunt, the patient developed progressive hiccups and stridor leading to respiratory failure requiring intubation. Ocular pendular nystagmus and palatal myoclonus at 3 Hz was observed. Flexible nasendoscopy (FNE) demonstrated bilateral tonic adduction of the vocal folds with 3 Hz coarse supraglottic, pharyngeal and palatal rhythmic myoclonus. MRI imaging demonstrated T2 hyperintensity within the bilateral inferior olivary nuclei consistent with stage 3 radiological HOD. CONCLUSIONS Dystonia is a rarely reported phenomenon in HOD but is not unexpected with the inferior olivary nucleus implicated in dystonic disorders. We report the association of HOD with bilateral vocal cord adductor dystonia, a potentially life threatening condition.
Collapse
Affiliation(s)
- Mitchell Lycett
- Department of Neurology, Royal North Shore Hospital, Reserve Road, St Leonards, New South Wales, 2065, Australia
| | - Cathy Kexin Cui
- Department of Neurology, Royal North Shore Hospital, Reserve Road, St Leonards, New South Wales, 2065, Australia
| | - Dijana Dragicevich
- Department of Speech Pathology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Roger Harris
- Department of Intensive Care Medicine, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Karl Ng
- Department of Neurology, Royal North Shore Hospital, Reserve Road, St Leonards, New South Wales, 2065, Australia.
| |
Collapse
|
31
|
Abstract
The generation of an internal body model and its continuous update is essential in sensorimotor control. Although known to rely on proprioceptive sensory feedback, the underlying mechanism that transforms this sensory feedback into a dynamic body percept remains poorly understood. However, advances in the development of genetic tools for proprioceptive circuit elements, including the sensory receptors, are beginning to offer new and unprecedented leverage to dissect the central pathways responsible for proprioceptive encoding. Simultaneously, new data derived through emerging bionic neural machine-interface technologies reveal clues regarding the relative importance of kinesthetic sensory feedback and insights into the functional proprioceptive substrates that underlie natural motor behaviors.
Collapse
Affiliation(s)
- Paul D Marasco
- Laboratory for Bionic Integration, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA;
- Charles Shor Epilepsy Center, Cleveland Clinic, Cleveland, Ohio, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - Joriene C de Nooij
- Department of Neurology and the Columbia University Motor Neuron Center, Columbia University Medical Center, New York, NY, USA;
| |
Collapse
|
32
|
Leon LES, Sillitoe RV. Disrupted sleep in dystonia depends on cerebellar function but not motor symptoms in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527916. [PMID: 36798256 PMCID: PMC9934608 DOI: 10.1101/2023.02.09.527916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Although dystonia is the third most common movement disorder, patients often also experience debilitating nonmotor defects including impaired sleep. The cerebellum is a central component of a "dystonia network" that plays various roles in sleep regulation. Importantly, the primary driver of sleep impairments in dystonia remains poorly understood. The cerebellum, along with other nodes in the motor circuit, could disrupt sleep. However, it is unclear how the cerebellum might alter sleep and mobility. To disentangle the impact of cerebellar dysfunction on motion and sleep, we generated two mouse genetic models of dystonia that have overlapping cerebellar circuit miswiring but show differing motor phenotype severity: Ptf1a Cre ;Vglut2 fx/fx and Pdx1 Cre ;Vglut2 fx/fx mice. In both models, excitatory climbing fiber to Purkinje cell neurotransmission is blocked, but only the Ptf1a Cre ;Vglut2 fx/fx mice have severe twisting. Using in vivo ECoG and EMG recordings we found that both mutants spend greater time awake and in NREM sleep at the expense of REM sleep. The increase in awake time is driven by longer awake bouts rather than an increase in bout number. We also found a longer latency to reach REM in both mutants, which is similar to what is reported in human dystonia. We uncovered independent but parallel roles for cerebellar circuit dysfunction and motor defects in promoting sleep quality versus posture impairments in dystonia.
Collapse
|
33
|
Beckinghausen J, Donofrio SG, Lin T, Miterko LN, White JJ, Lackey EP, Sillitoe RV. Deep Brain Stimulation of the Interposed Cerebellar Nuclei in a Conditional Genetic Mouse Model with Dystonia. ADVANCES IN NEUROBIOLOGY 2023; 31:93-117. [PMID: 37338698 DOI: 10.1007/978-3-031-26220-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Dystonia is a neurological disease that is currently ranked as the third most common motor disorder. Patients exhibit repetitive and sometimes sustained muscle contractions that cause limb and body twisting and abnormal postures that impair movement. Deep brain stimulation (DBS) of the basal ganglia and thalamus can be used to improve motor function when other treatment options fail. Recently, the cerebellum has garnered interest as a DBS target for treating dystonia and other motor disorders. Here, we describe a procedure for targeting DBS electrodes to the interposed cerebellar nuclei to correct motor dysfunction in a mouse model with dystonia. Targeting cerebellar outflow pathways with neuromodulation opens new possibilities for using the expansive connectivity of the cerebellum to treat motor and non-motor diseases.
Collapse
Affiliation(s)
- Jaclyn Beckinghausen
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA
| | - Sarah G Donofrio
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA
| | - Tao Lin
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA
| | - Lauren N Miterko
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - Joshua J White
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA
| | - Elizabeth P Lackey
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA
| | - Roy V Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA.
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
34
|
Manto M, Mitoma H. Cerebellum: From the identification of the cerebellar motor syndrome to the internal models. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:159-174. [PMID: 37620068 DOI: 10.1016/b978-0-323-98817-9.00024-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Cerebellar circuitry is topographically arranged in closed loops with the cerebral cortex. The three cornerstones of clinical ataxia have emerged from studies on connectional anatomy and from clinical/neuropsychological observations, leading to the definition of clinical syndromes encountered in daily practice: (a) the cerebellar motor syndrome (CMS), (b) the vestibulocerebellar syndrome (VCS), and (c) the cerebellar cognitive affective syndrome/Schmahmann syndrome (CCAS/SS). These syndromes are either isolated or coexist, depending on the underlying pathological process and its degree of extension within the cerebellum. Dysmetria is the core feature of cerebellar deficits, encompassing motor dysmetria (hypermetria, hypometria) in CMS, oculomotor dysmetria in VCS, and dysmetria of thought in CCAS/SS. The leading hypothesis is that dysmetria results from errors in building or maintaining internal models, which are inherent to predictive behavior. Errors in prediction would lead to clumsiness and incoordination of limbs, oculomotor impairments, and aberrant cognitive/affective behavior. The cerebellum is currently viewed as a learning machine enriched with multiple plasticity mechanisms, allowing the permanent adaptation to the external world by generating and maintaining predictive operations, from motor to cognitive, affective, emotional, and social operations essential for daily human life.
Collapse
Affiliation(s)
- Mario Manto
- Unité des Ataxies Cérébelleuses, Service de Neurologie, CHU-Charleroi, Charleroi, Belgium; Service des Neurosciences, Université de Mons, Mons, Belgium.
| | - Hiroshi Mitoma
- Department of Medical Education, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
35
|
Brown AM, Lackey EP, Salazar Leon LE, Rey Hipolito AG, Beckinghausen J, Lin T, Sillitoe RV. Electromyography as a Method for Distinguishing Dystonia in Mice. ADVANCES IN NEUROBIOLOGY 2023; 31:71-91. [PMID: 37338697 DOI: 10.1007/978-3-031-26220-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Electromyography (EMG) methods allow quantitative analyses of motor function. The techniques include intramuscular recordings that are performed in vivo. However, recording muscle activity in freely moving mice, particularly in models of motor disease, often creates challenges that prevent the acquisition of clean signals. Recording preparations must be stable enough for the experimenter to collect an adequate number of signals for statistical analyses. Instability results in a low signal-to-noise ratio that prohibits proper isolation of EMG signals from the target muscle during the behavior of interest. Such insufficient isolation prevents the analysis of full electrical potential waveforms. In this case, resolving the shape of a waveform to differentiate individual spikes and bursts of muscle activity can be difficult. A common source of instability is an inadequate surgery. Poor surgical techniques cause blood loss, tissue damage, poor healing, encumbered movement, and unstable implantation of the electrodes. Here, we describe an optimized surgical procedure that ensures electrode stability for in vivo muscle recordings. We implement our technique to obtain recordings from agonist and antagonist muscle pairs in the hindlimbs of freely moving adult mice. We validate the stability of our method by holding EMG recordings during dystonic behavior. Our approach is ideal for studying normal and abnormal motor function in actively behaving mice and valuable for recording intramuscular activity when considerable motion is expected.
Collapse
Affiliation(s)
- Amanda M Brown
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Elizabeth P Lackey
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Luis E Salazar Leon
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Alejandro G Rey Hipolito
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Jaclyn Beckinghausen
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Tao Lin
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Roy V Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
36
|
Liu B, Mao Z, Cui Z, Ling Z, Xu X, He K, Cui M, Feng Z, Yu X, Zhang Y. Cerebellar gray matter alterations predict deep brain stimulation outcomes in Meige syndrome. Neuroimage Clin 2023; 37:103316. [PMID: 36610311 PMCID: PMC9827385 DOI: 10.1016/j.nicl.2023.103316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/21/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
BACKGROUND The physiopathologic mechanism of Meige syndrome (MS) has not been clarified, and neuroimaging studies centering on cerebellar changes in MS are scarce. Moreover, even though deep brain stimulation (DBS) of the subthalamic nucleus (STN) has been recognized as an effective surgical treatment for MS, there has been no reliable biomarker to predict its efficacy. OBJECTIVE To characterize the volumetric alterations of gray matter (GM) in the cerebellum in MS and to identify GM measurements related to a good STN-DBS outcome. METHODS We used voxel-based morphometry and lobule-based morphometry to compare the regional and lobular GM differences in the cerebellum between 47 MS patients and 52 normal human controls (HCs), as well as between 31 DBS responders and 10 DBS non-responders. Both volumetric analyses were achieved using the Spatially Unbiased Infratentorial Toolbox (SUIT). Further, we performed partial correlation analyses to probe the relationship between the cerebellar GM changes and clinical scores. Finally, we plotted the receiver operating characteristic (ROC) curve to select biomarkers for MS diagnosis and DBS outcomes prediction. RESULTS Compared to HCs, MS patients had GM atrophy in lobule Crus I, lobule VI, lobule VIIb, lobule VIIIa, and lobule VIIIb. Compared to DBS responders, DBS non-responders had lower GM volume in the left lobule VIIIb. Moreover, partial correlation analyses revealed a positive relationship between the GM volume of the significant regions/lobules and the symptom improvement rate after DBS surgery. ROC analyses demonstrated that the GM volume of the significant cluster in the left lobule VIIIb could not only distinguish MS patients from HCs but also predict the outcomes of STN-DBS surgery with high accuracy. CONCLUSION MS patients display bilateral GM shrinkage in the cerebellum relative to HCs. Regional GM volume of the left lobule VIIIb can be a reliable biomarker for MS diagnosis and DBS outcomes prediction.
Collapse
Affiliation(s)
- Bin Liu
- Medical School of Chinese PLA, Beijing, PR China; Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China
| | - Zhiqi Mao
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China
| | - Zhiqiang Cui
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China
| | - Zhipei Ling
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China
| | - Xin Xu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China
| | - Kunyu He
- Medical School of Chinese PLA, Beijing, PR China; Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China
| | - Mengchu Cui
- Medical School of Chinese PLA, Beijing, PR China; Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China
| | - Zhebin Feng
- Medical School of Chinese PLA, Beijing, PR China; Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China
| | - Xinguang Yu
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China; Neurosurgery Institute, Chinese PLA General Hospital, Beijing, PR China.
| | - Yanyang Zhang
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing, PR China; Neurosurgery Institute, Chinese PLA General Hospital, Beijing, PR China.
| |
Collapse
|
37
|
Zhou J, Van der Heijden ME, Salazar Leon LE, Lin T, Miterko LN, Kizek DJ, Perez RM, Pavešković M, Brown AM, Sillitoe RV. Propranolol Modulates Cerebellar Circuit Activity and Reduces Tremor. Cells 2022; 11:cells11233889. [PMID: 36497147 PMCID: PMC9740691 DOI: 10.3390/cells11233889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/10/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Tremor is the most common movement disorder. Several drugs reduce tremor severity, but no cures are available. Propranolol, a β-adrenergic receptor blocker, is the leading treatment for tremor. However, the in vivo circuit mechanisms by which propranolol decreases tremor remain unclear. Here, we test whether propranolol modulates activity in the cerebellum, a key node in the tremor network. We investigated the effects of propranolol in healthy control mice and Car8wdl/wdl mice, which exhibit pathophysiological tremor and ataxia due to cerebellar dysfunction. Propranolol reduced physiological tremor in control mice and reduced pathophysiological tremor in Car8wdl/wdl mice to control levels. Open field and footprinting assays showed that propranolol did not correct ataxia in Car8wdl/wdl mice. In vivo recordings in awake mice revealed that propranolol modulates the spiking activity of control and Car8wdl/wdl Purkinje cells. Recordings in cerebellar nuclei neurons, the targets of Purkinje cells, also revealed altered activity in propranolol-treated control and Car8wdl/wdl mice. Next, we tested whether propranolol reduces tremor through β1 and β2 adrenergic receptors. Propranolol did not change tremor amplitude or cerebellar nuclei activity in β1 and β2 null mice or Car8wdl/wdl mice lacking β1 and β2 receptor function. These data show that propranolol can modulate cerebellar circuit activity through β-adrenergic receptors and may contribute to tremor therapeutics.
Collapse
Affiliation(s)
- Joy Zhou
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meike E. Van der Heijden
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Luis E. Salazar Leon
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tao Lin
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Lauren N. Miterko
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dominic J. Kizek
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Ross M. Perez
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matea Pavešković
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Amanda M. Brown
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Roy V. Sillitoe
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-832-824-8913
| |
Collapse
|
38
|
van der Heijden ME, Brown AM, Sillitoe RV. Influence of data sampling methods on the representation of neural spiking activity in vivo. iScience 2022; 25:105429. [PMID: 36388953 PMCID: PMC9641233 DOI: 10.1016/j.isci.2022.105429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/06/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
In vivo single-unit recordings distinguish the basal spiking properties of neurons in different experimental settings and disease states. Here, we examined over 300 spike trains recorded from Purkinje cells and cerebellar nuclei neurons to test whether data sampling approaches influence the extraction of rich descriptors of firing properties. Our analyses included neurons recorded in awake and anesthetized control mice, and disease models of ataxia, dystonia, and tremor. We find that recording duration circumscribes overall representations of firing rate and pattern. Notably, shorter recording durations skew estimates for global firing rate variability toward lower values. We also find that only some populations of neurons in the same mouse are more similar to each other than to neurons recorded in different mice. These data reveal that recording duration and approach are primary considerations when interpreting task-independent single neuron firing properties. If not accounted for, group differences may be concealed or exaggerated.
Collapse
Affiliation(s)
- Meike E. van der Heijden
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| | - Amanda M. Brown
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| | - Roy V. Sillitoe
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
39
|
Masoli S, Rizza MF, Tognolina M, Prestori F, D’Angelo E. Computational models of neurotransmission at cerebellar synapses unveil the impact on network computation. Front Comput Neurosci 2022; 16:1006989. [PMID: 36387305 PMCID: PMC9649760 DOI: 10.3389/fncom.2022.1006989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
The neuroscientific field benefits from the conjoint evolution of experimental and computational techniques, allowing for the reconstruction and simulation of complex models of neurons and synapses. Chemical synapses are characterized by presynaptic vesicle cycling, neurotransmitter diffusion, and postsynaptic receptor activation, which eventually lead to postsynaptic currents and subsequent membrane potential changes. These mechanisms have been accurately modeled for different synapses and receptor types (AMPA, NMDA, and GABA) of the cerebellar cortical network, allowing simulation of their impact on computation. Of special relevance is short-term synaptic plasticity, which generates spatiotemporal filtering in local microcircuits and controls burst transmission and information flow through the network. Here, we present how data-driven computational models recapitulate the properties of neurotransmission at cerebellar synapses. The simulation of microcircuit models is starting to reveal how diverse synaptic mechanisms shape the spatiotemporal profiles of circuit activity and computation.
Collapse
Affiliation(s)
- Stefano Masoli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | | | | | - Francesca Prestori
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- IRCCS Mondino Foundation, Brain Connectivity Center, Pavia, Italy
| |
Collapse
|
40
|
Salazar Leon LE, Sillitoe RV. Potential Interactions Between Cerebellar Dysfunction and Sleep Disturbances in Dystonia. DYSTONIA 2022; 1. [PMID: 37065094 PMCID: PMC10099477 DOI: 10.3389/dyst.2022.10691] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Dystonia is the third most common movement disorder. It causes debilitating twisting postures that are accompanied by repetitive and sometimes intermittent co- or over-contractions of agonist and antagonist muscles. Historically diagnosed as a basal ganglia disorder, dystonia is increasingly considered a network disorder involving various brain regions including the cerebellum. In certain etiologies of dystonia, aberrant motor activity is generated in the cerebellum and the abnormal signals then propagate through a “dystonia circuit” that includes the thalamus, basal ganglia, and cerebral cortex. Importantly, it has been reported that non-motor defects can accompany the motor symptoms; while their severity is not always correlated, it is hypothesized that common pathways may nevertheless be disrupted. In particular, circadian dysfunction and disordered sleep are common non-motor patient complaints in dystonia. Given recent evidence suggesting that the cerebellum contains a circadian oscillator, displays sleep-stage-specific neuronal activity, and sends robust long-range projections to several subcortical regions involved in circadian rhythm regulation, disordered sleep in dystonia may result from cerebellum-mediated dysfunction of the dystonia circuit. Here, we review the evidence linking dystonia, cerebellar network dysfunction, and cerebellar involvement in sleep. Together, these ideas may form the basis for the development of improved pharmacological and surgical interventions that could take advantage of cerebellar circuitry to restore normal motor function as well as non-motor (sleep) behaviors in dystonia.
Collapse
Affiliation(s)
- Luis E. Salazar Leon
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, 77030, USA
| | - Roy V. Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, 77030, USA
- Address correspondence to: Dr. Roy V. Sillitoe, Tel: 832-824-8913, Fax: 832-825-1251,
| |
Collapse
|
41
|
Rodichkin AN, Edler MK, McGlothan JL, Guilarte TR. Pathophysiological studies of aging Slc39a14 knockout mice to assess the progression of manganese-induced dystonia-parkinsonism. Neurotoxicology 2022; 93:92-102. [PMID: 36152728 DOI: 10.1016/j.neuro.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Over the last decade, several clinical reports have outlined cases of early-onset manganese (Mn)-induced dystonia-parkinsonism, resulting from loss of function mutations of the Mn transporter gene SLC39A14. Previously, we have performed characterization of the behavioral, neurochemical, and neuropathological changes in 60-day old (PN60) Slc39a14-knockout (KO) murine model of the human disease. Here, we extend our studies to aging Slc39a14-KO mice to assess the progression of the disease. Our results indicate that 365-day old (PN365) Slc39a14-KO mice present with markedly elevated blood and brain Mn levels, similar to those found in the PN60 mice and representative of the human cases of the disease. Furthermore, aging Slc39a14-KO mice consistently manifest a hypoactive and dystonic behavioral deficits, similar to the PN60 animals, suggesting that the behavioral changes are established early in life without further age-associated deterioration. Neurochemical, neuropathological, and functional assessment of the dopaminergic system of the basal ganglia revealed absence of neurodegenerative changes of dopamine (DA) neurons in the substantia nigra pars compacta (SNc), with no changes in DA or metabolite concentrations in the striatum of Slc39a14-KO mice relative to wildtype (WT). Similar to the PN60 animals, aging Slc39a14-KO mice expressed a marked inhibition of potassium-stimulated DA release in the striatum. Together our findings indicate that the pathophysiological changes observed in the basal ganglia of aging Slc39a14-KO animals are similar to those at PN60 and aging does not have a significant effect on these parameters.
Collapse
Affiliation(s)
- Alexander N Rodichkin
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States.
| | - Melissa K Edler
- Department of Anthropology and Brain Health Research Institute, Kent State University, Kent, OH 44242, United States.
| | - Jennifer L McGlothan
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States.
| | - Tomás R Guilarte
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States.
| |
Collapse
|
42
|
Asthana P, Kumar G, Milanowski LM, Au NPB, Chan SC, Huang J, Feng H, Kwan KM, He J, Chan KWY, Wszolek ZK, Ma CHE. Cerebellar glutamatergic system impacts spontaneous motor recovery by regulating Gria1 expression. NPJ Regen Med 2022; 7:45. [PMID: 36064798 PMCID: PMC9445039 DOI: 10.1038/s41536-022-00243-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
Peripheral nerve injury (PNI) often results in spontaneous motor recovery; however, how disrupted cerebellar circuitry affects PNI-associated motor recovery is unknown. Here, we demonstrated disrupted cerebellar circuitry and poor motor recovery in ataxia mice after PNI. This effect was mimicked by deep cerebellar nuclei (DCN) lesion, but not by damaging non-motor area hippocampus. By restoring cerebellar circuitry through DCN stimulation, and reversal of neurotransmitter imbalance using baclofen, ataxia mice achieve full motor recovery after PNI. Mechanistically, elevated glutamate-glutamine level was detected in DCN of ataxia mice by magnetic resonance spectroscopy. Transcriptomic study revealed that Gria1, an ionotropic glutamate receptor, was upregulated in DCN of control mice but failed to be upregulated in ataxia mice after sciatic nerve crush. AAV-mediated overexpression of Gria1 in DCN rescued motor deficits of ataxia mice after PNI. Finally, we found a correlative decrease in human GRIA1 mRNA expression in the cerebellum of patients with ataxia-telangiectasia and spinocerebellar ataxia type 6 patient iPSC-derived Purkinje cells, pointing to the clinical relevance of glutamatergic system. By conducting a large-scale analysis of 9,655,320 patients with ataxia, they failed to recover from carpal tunnel decompression surgery and tibial neuropathy, while aged-match non-ataxia patients fully recovered. Our results provide insight into cerebellar disorders and motor deficits after PNI.
Collapse
Affiliation(s)
- Pallavi Asthana
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Lukasz M Milanowski
- Department of Neurology, Mayo Clinic, Jacksonville, USA.,Department of Neurology, Faculty of Health Science, Medical University of Warsaw, Warsaw, Poland
| | - Ngan Pan Bennett Au
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Siu Chung Chan
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Hemin Feng
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Kin Ming Kwan
- School of Life Sciences, Center for Cell and Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Jufang He
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR
| | - Kannie Wai Yan Chan
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, USA
| | | | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Hong Kong SAR.
| |
Collapse
|
43
|
Ponce GV, Klaus J, Schutter DJLG. A Brief History of Cerebellar Neurostimulation. CEREBELLUM (LONDON, ENGLAND) 2022; 21:715-730. [PMID: 34403075 PMCID: PMC9325826 DOI: 10.1007/s12311-021-01310-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 12/28/2022]
Abstract
The first attempts at using electric stimulation to study human brain functions followed the experiments of Luigi Galvani and Giovanni Aldini on animal electricity during the eighteenth century. Since then, the cerebellum has been among the areas that have been studied by invasive and non-invasive forms of electrical and magnetic stimulation. During the nineteenth century, animal experiments were conducted to map the motor-related regions of cerebellar cortex by means of direct electric stimulation. As electric stimulation research on the cerebellum moved into the twentieth century, systematic research of electric cerebellar stimulation led to a better understanding of its effects and mechanism of action. In addition, the clinical potential of cerebellar stimulation in the treatment of motor diseases started to be explored. With the introduction of transcranial electric and magnetic stimulation, cerebellar research moved to non-invasive techniques. During the twenty-first century, following on groundbreaking research that linked the cerebellum to non-motor functions, non-invasive techniques have facilitated research into different aspects of cerebellar functioning. The present review provides a brief historical account of cerebellar neurostimulation and discusses current challenges and future direction in this field of research.
Collapse
Affiliation(s)
- Gustavo V Ponce
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Heidelberglaan 1, 3584CS, Utrecht, The Netherlands
| | - Jana Klaus
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Heidelberglaan 1, 3584CS, Utrecht, The Netherlands
| | - Dennis J L G Schutter
- Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Heidelberglaan 1, 3584CS, Utrecht, The Netherlands.
| |
Collapse
|
44
|
Bauer S, van Wingerden N, Jacobs T, van der Horst A, Zhai P, Betting JHLF, Strydis C, White JJ, De Zeeuw CI, Romano V. Purkinje Cell Activity Resonation Generates Rhythmic Behaviors at the Preferred Frequency of 8 Hz. Biomedicines 2022; 10:biomedicines10081831. [PMID: 36009378 PMCID: PMC9404806 DOI: 10.3390/biomedicines10081831] [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: 06/30/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 12/01/2022] Open
Abstract
Neural activity exhibits oscillations, bursts, and resonance, enhancing responsiveness at preferential frequencies. For example, theta-frequency bursting and resonance in granule cells facilitate synaptic transmission and plasticity mechanisms at the input stage of the cerebellar cortex. However, whether theta-frequency bursting of Purkinje cells is involved in generating rhythmic behavior has remained neglected. We recorded and optogenetically modulated the simple and complex spike activity of Purkinje cells while monitoring whisker movements with a high-speed camera of awake, head-fixed mice. During spontaneous whisking, both simple spike activity and whisker movement exhibit peaks within the theta band. Eliciting either simple or complex spikes at frequencies ranging from 0.5 to 28 Hz, we found that 8 Hz is the preferred frequency around which the largest movement is induced. Interestingly, oscillatory whisker movements at 8 Hz were also generated when simple spike bursting was induced at 2 and 4 Hz, but never via climbing fiber stimulation. These results indicate that 8 Hz is the resonant frequency at which the cerebellar-whisker circuitry produces rhythmic whisking.
Collapse
Affiliation(s)
- Staf Bauer
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
| | - Nathalie van Wingerden
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
| | - Thomas Jacobs
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
| | - Annabel van der Horst
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
| | - Peipei Zhai
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
| | - Jan-Harm L. F. Betting
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
| | - Christos Strydis
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
- Department of Quantum & Computing Engineering, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Joshua J. White
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
- Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands
| | - Vincenzo Romano
- Department of Neuroscience, Erasmus MC, 3015 GD Rotterdam, The Netherlands; (S.B.); (N.v.W.); (T.J.); (A.v.d.H.); (P.Z.); (J.-H.L.F.B.); (C.S.); (J.J.W.); (C.I.D.Z.)
- Correspondence:
| |
Collapse
|
45
|
Noseda R. Cerebro-Cerebellar Networks in Migraine Symptoms and Headache. FRONTIERS IN PAIN RESEARCH 2022; 3:940923. [PMID: 35910262 PMCID: PMC9326053 DOI: 10.3389/fpain.2022.940923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
The cerebellum is associated with the biology of migraine in a variety of ways. Clinically, symptoms such as fatigue, motor weakness, vertigo, dizziness, difficulty concentrating and finding words, nausea, and visual disturbances are common in different types of migraine. The neural basis of these symptoms is complex, not completely known, and likely involve activation of both specific and shared circuits throughout the brain. Posterior circulation stroke, or neurosurgical removal of posterior fossa tumors, as well as anatomical tract tracing in animals, provided the first insights to theorize about cerebellar functions. Nowadays, with the addition of functional imaging, much progress has been done on cerebellar structure and function in health and disease, and, as a consequence, the theories refined. Accordingly, the cerebellum may be useful but not necessary for the execution of motor, sensory or cognitive tasks, but, rather, would participate as an efficiency facilitator of neurologic functions by improving speed and skill in performance of tasks produced by the cerebral area to which it is reciprocally connected. At the subcortical level, critical regions in these processes are the basal ganglia and thalamic nuclei. Altogether, a modulatory role of the cerebellum over multiple brain regions appears compelling, mainly by considering the complexity of its reciprocal connections to common neural networks involved in motor, vestibular, cognitive, affective, sensory, and autonomic processing—all functions affected at different phases and degrees across the migraine spectrum. Despite the many associations between cerebellum and migraine, it is not known whether this structure contributes to migraine initiation, symptoms generation or headache. Specific cerebellar dysfunction via genetically driven excitatory/inhibitory imbalances, oligemia and/or increased risk to white matter lesions has been proposed as a critical contributor to migraine pathogenesis. Therefore, given that neural projections and functions of many brainstem, midbrain and forebrain areas are shared between the cerebellum and migraine trigeminovascular pathways, this review will provide a synopsis on cerebellar structure and function, its role in trigeminal pain, and an updated overview of relevant clinical and preclinical literature on the potential role of cerebellar networks in migraine pathophysiology.
Collapse
Affiliation(s)
- Rodrigo Noseda
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- *Correspondence: Rodrigo Noseda
| |
Collapse
|
46
|
Geminiani A, Mockevicius A, D'Angelo E, Casellato C. Cerebellum involvement in dystonia: insights from a spiking neural network model during associative learning. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:5132-5135. [PMID: 36086302 DOI: 10.1109/embc48229.2022.9871205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Dystonia is a neurological movement disorder characterized by twisting and repetitive movements or abnormal fixed postures. This complex brain disease has usually been associated with damages to the Basal Ganglia. However, recent studies point out the potential role of the cerebellum. Indeed, motor learning is impaired in dystonic patients, e.g. during eyeblink classical conditioning, a typical cerebellum-driven associative learning protocol, and rodents with local cerebellar damages exhibit dystonic movements. Alterations in the olivocerebellar circuit connectivity have been identified as a potential neural substrate of dystonia. Here, we investigated this hypothesis through simulations of eyeblink conditioning driven by a realistic spiking model of the cerebellum. The pathological model was generated by decreasing the signal transmission from the Inferior Olive to cerebellar cortex, as observed in animal experiments. The model was able to reproduce a reduced acquisition of eyeblink motor responses, with also an unproper timing. Indeed, this pathway is fundamental to drive cerebellar cortical plasticity, which is the basis of cerebellum-driven motor learning. Exploring different levels of damage, the model predicted the possible amount of underlying impairment associated with the misbehavior observed in patients. Simulations of other debated lesions reported in mouse models of dystonia will be run to investigate the cerebellar involvement in different types of dystonia. Indeed, the eyeblink conditioning phenotype could be used to discriminate between them, identifying specific deficits in the generation of motor responses. Future studies will also include simulations of pharmacological or deep brain stimulation treatments targeting the cerebellum, to predict their impact in improving symptoms.
Collapse
|
47
|
Geminiani A, Mockevičius A, D’Angelo E, Casellato C. Cerebellum Involvement in Dystonia During Associative Motor Learning: Insights From a Data-Driven Spiking Network Model. Front Syst Neurosci 2022; 16:919761. [PMID: 35782305 PMCID: PMC9243665 DOI: 10.3389/fnsys.2022.919761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Dystonia is a movement disorder characterized by sustained or intermittent muscle contractions causing abnormal, often repetitive movements, postures, or both. Although dystonia is traditionally associated with basal ganglia dysfunction, recent evidence has been pointing to a role of the cerebellum, a brain area involved in motor control and learning. Cerebellar abnormalities have been correlated with dystonia but their potential causative role remains elusive. Here, we simulated the cerebellar input-output relationship with high-resolution computational modeling. We used a data-driven cerebellar Spiking Neural Network and simulated a cerebellum-driven associative learning task, Eye-Blink Classical Conditioning (EBCC), which is characteristically altered in relation to cerebellar lesions in several pathologies. In control simulations, input stimuli entrained characteristic network dynamics and induced synaptic plasticity along task repetitions, causing a progressive spike suppression in Purkinje cells with consequent facilitation of deep cerebellar nuclei cells. These neuronal processes caused a progressive acquisition of eyelid Conditioned Responses (CRs). Then, we modified structural or functional local neural features in the network reproducing alterations reported in dystonic mice. Either reduced olivocerebellar input or aberrant Purkinje cell burst-firing resulted in abnormal learning curves imitating the dysfunctional EBCC motor responses (in terms of CR amount and timing) of dystonic mice. These behavioral deficits might be due to altered temporal processing of sensorimotor information and uncoordinated control of muscle contractions. Conversely, an imbalance of excitatory and inhibitory synaptic densities on Purkinje cells did not reflect into significant EBCC deficit. The present work suggests that only certain types of alterations, including reduced olivocerebellar input and aberrant PC burst-firing, are compatible with the EBCC changes observed in dystonia, indicating that some cerebellar lesions can have a causative role in the pathogenesis of symptoms.
Collapse
Affiliation(s)
- Alice Geminiani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Aurimas Mockevičius
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Brain Connectivity Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Claudia Casellato
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
48
|
Kumar G, Asthana P, Yung WH, Kwan KM, Tin C, Ma CHE. Deep Brain Stimulation of the Interposed Nucleus Reverses Motor Deficits and Stimulates Production of Anti-inflammatory Cytokines in Ataxia Mice. Mol Neurobiol 2022; 59:4578-4592. [PMID: 35581519 DOI: 10.1007/s12035-022-02872-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/05/2022] [Indexed: 11/24/2022]
Abstract
Cerebellum is one of the major targets of autoimmunity and cerebellar damage that leads to ataxia characterized by the loss of fine motor coordination and balance, with no treatment available. Deep brain stimulation (DBS) could be a promising treatment for ataxia but has not been extensively investigated. Here, our study aims to investigate the use of interposed nucleus of deep cerebellar nuclei (IN-DCN) for ataxia. We first characterized ataxia-related motor symptom of a Purkinje cell (PC)-specific LIM homeobox (Lhx)1 and Lhx5 conditional double knockout mice by motor coordination tests, and spontaneous electromyogram (EMG) recording. To validate IN-DCN as a target for DBS, in vivo local field potential (LFP) multielectrode array recording of IN-DCN revealed abnormal LFP amplitude surges in PCs. By synchronizing the EMG and IN-DCN recordings (neurospike and LFP) with high-speed video recordings, ataxia mice showed poorly coordinated movements associated with low EMG amplitude and aberrant IN-DCN neural firing. To optimize IN-DCN-DBS for ataxia, we tested DBS parameters from low (30 Hz) to high stimulation frequency (130 or 150 Hz), and systematically varied pulse width values (60 or 80 µs) to maximize motor symptom control in ataxia mice. The optimal IN-DCN-DBS parameter reversed motor deficits in ataxia mice as detected by animal behavioral tests and EMG recording. Mechanistically, cytokine array analysis revealed that anti-inflammatory cytokines such as interleukin (IL)-13 and IL-4 were upregulated after IN-DCN-DBS, which play key roles in neural excitability. As such, we show that IN-DCN-DBS is a promising treatment for ataxia and possibly other movement disorders alike.
Collapse
Affiliation(s)
- Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Hong Kong SAR, China
| | - Pallavi Asthana
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Hong Kong SAR, China
| | - Wing Ho Yung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, China
| | - Kin Ming Kwan
- School of Life Sciences, Center for Cell and Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, China
| | - Chung Tin
- Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Hong Kong, SAR, China
| | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Hong Kong SAR, China.
| |
Collapse
|
49
|
Dorgans K, Guo D, Kurima K, Wickens J, Uusisaari MY. Designing AAV Vectors for Monitoring the Subtle Calcium Fluctuations of Inferior Olive Network in vivo. Front Cell Neurosci 2022; 16:825056. [PMID: 35573836 PMCID: PMC9093741 DOI: 10.3389/fncel.2022.825056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Adeno-associated viral (AAV) vectors, used as vehicles for gene transfer into the brain, are a versatile and powerful tool of modern neuroscience that allow identifying specific neuronal populations, monitoring and modulating their activity. For consistent and reproducible results, the AAV vectors must be engineered so that they reliably and accurately target cell populations. Furthermore, transgene expression must be adjusted to sufficient and safe levels compatible with the physiology of studied cells. We undertook the effort to identify and validate an AAV vector that could be utilized for researching the inferior olivary (IO) nucleus, a structure gating critical timing-related signals to the cerebellum. By means of systematic construct generation and quantitative expression profiling, we succeeded in creating a viral tool for specific and strong transfection of the IO neurons without adverse effects on their physiology. The potential of these tools is demonstrated by expressing the calcium sensor GCaMP6s in adult mouse IO neurons. We could monitor subtle calcium fluctuations underlying two signatures of intrinsic IO activity: the subthreshold oscillations (STOs) and the variable-duration action potential waveforms both in-vitro and in-vivo. Further, we show that the expression levels of GCaMP6s allowing such recordings are compatible with the delicate calcium-based dynamics of IO neurons, inviting future work into the network dynamics of the olivo-cerebellar system in behaving animals.
Collapse
Affiliation(s)
- Kevin Dorgans
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Da Guo
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Kiyoto Kurima
- Neurobiology Research Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Jeff Wickens
- Neurobiology Research Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Marylka Yoe Uusisaari
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- *Correspondence: Marylka Yoe Uusisaari
| |
Collapse
|
50
|
Romano V, Zhai P, van der Horst A, Mazza R, Jacobs T, Bauer S, Wang X, White JJ, De Zeeuw CI. Olivocerebellar control of movement symmetry. Curr Biol 2022; 32:654-670.e4. [PMID: 35016009 DOI: 10.1016/j.cub.2021.12.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/26/2021] [Accepted: 12/08/2021] [Indexed: 01/02/2023]
Abstract
Coordination of bilateral movements is essential for a large variety of animal behaviors. The olivocerebellar system is critical for the control of movement, but its role in bilateral coordination has yet to be elucidated. Here, we examined whether Purkinje cells encode and influence synchronicity of left-right whisker movements. We found that complex spike activity is correlated with a prominent left-right symmetry of spontaneous whisker movements within parts, but not all, of Crus1 and Crus2. Optogenetic stimulation of climbing fibers in the areas with high and low correlations resulted in symmetric and asymmetric whisker movements, respectively. Moreover, when simple spike frequency prior to the complex spike was higher, the complex spike-related symmetric whisker protractions were larger. This finding alludes to a role for rebound activity in the cerebellar nuclei, which indeed turned out to be enhanced during symmetric protractions. Tracer injections suggest that regions associated with symmetric whisker movements are anatomically connected to the contralateral cerebellar hemisphere. Together, these data point toward the existence of modules on both sides of the cerebellar cortex that can differentially promote or reduce the symmetry of left and right movements in a context-dependent fashion.
Collapse
Affiliation(s)
- Vincenzo Romano
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands.
| | - Peipei Zhai
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | - Roberta Mazza
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Thomas Jacobs
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Staf Bauer
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Xiaolu Wang
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Joshua J White
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - C I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands; Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, the Netherlands.
| |
Collapse
|