1
|
Hsiao CT, Fu SJ, Cheng KM, Lo H, Tang CY, Chan CC, Jeng CJ. Restoration of Shal/K V4 proteostasis and motor function in a Drosophila model of spinocerebellar ataxia type 19/22. Cell Mol Life Sci 2025; 82:181. [PMID: 40293501 PMCID: PMC12037467 DOI: 10.1007/s00018-025-05711-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025]
Abstract
Loss-of-function mutations in the human KCND3 gene encoding KV4.3 K+ channels are linked to the autosomal dominant neurodegenerative disease spinocerebellar ataxia type 19/22 (SCA19/22). Previous biophysical and biochemical analyses in vitro support the notion that the autosomal dominant inheritance pattern of SCA19/22 is associated with the dominant-negative effects of disease-causing KV4.3 mutants on proteostasis of their wild-type (WT) counterpart. Herein we aimed to explore whether the disease-causing mutants might perturb protein expression of endogenous KV4.3 channel in human cells, as well as contributing to in vivo pathomechanisms underlying motor impairments and neurodegeneration in an animal model of SCA19/22. Substantial reduction in human KV4.3 protein level was validated in skin fibroblasts derived from heterozygous SCA19/22 patients. Genetic knockdown of endogenous Shal, the fly ortholog of human KV4.3, in Drosophila led to locomotor impairment, ommatidia degeneration, and reduced brain cortex thickness, all of which was effectively ameliorated by transgenic expression of human KV4.3, but not KV1.1 K+ channel. Transgenic expression of SCA19/22-causing human KV4.3 mutants resulted in notable disruption of endogenous Shal proteostasis, locomotor function, and ommatidia morphology in Drosophila. Enhanced expression of the Drosophila molecular chaperones HSC70 and HSP83 in our fly model of SCA19/22 corrected Shal protein deficit, locomotor dysfunction, and neurodegeneration. Overexpression of Hsp90β also upregulated endogenous human KV4.3 protein level in patient-derived skin fibroblasts. Our findings highlight Drosophila as a suitable animal model for studying KV4.3 channelopathy in vivo, and accentuate a critical role of defective KV4.3 proteostasis in the pathogenesis of motor dysfunction and neurodegeneration in SCA19/22.
Collapse
Affiliation(s)
- Cheng-Tsung Hsiao
- Department of Neurology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Neurology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Ssu-Ju Fu
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Kai-Min Cheng
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Hsiang Lo
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Chih-Yung Tang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| | - Chih-Chiang Chan
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| | - Chung-Jiuan Jeng
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
2
|
Cornforth E, Schmahmann JD. Physical Therapy and Aminopyridine for Downbeat Nystagmus Syndrome: A Case Report. J Neurol Phys Ther 2025; 49:108-113. [PMID: 38898545 DOI: 10.1097/npt.0000000000000485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
BACKGROUND AND PURPOSE Individuals with downbeat nystagmus (DBN) syndrome present with DBN, dizziness, blurred vision, and unsteady gait. Pharmacological intervention with 4-aminopyridine (4-AP) may be effective in improving oculomotor function, but there is minimal evidence to date that it improves gait. This suggests the possible benefit of combining pharmacotherapy with physical therapy to maximize outcomes. This case report documents improvements in gait and balance after physical therapy and aminopyridine (AP) in an individual with DBN syndrome. CASE DESCRIPTION The patient was a 70-year-old man with a 4-year history of worsening dizziness and imbalance, diagnosed with DBN syndrome. He demonstrated impaired oculomotor function, dizziness, and imbalance, which resulted in falls and limited community ambulation. INTERVENTION The patient completed a customized, tapered course of physical therapy over 6 months. Outcome measures included the 10-meter walk test, the Timed Up and Go (TUG), the Dynamic Gait Index (DGI), and the modified clinical test of sensory integration and balance. OUTCOMES Improvements exceeding minimal detectable change were demonstrated on the TUG and the DGI. Gait speed on the 10-meter walk test did not change significantly, but the patient was able to use a cane to ambulate in the community and reported no further falls. DISCUSSION Controlled studies are needed to explore the potential for AP to augment physical therapy in people with DBN syndrome. Physical therapists are encouraged to communicate with referring medical providers about the use of AP as pharmacotherapy along with physical therapy for individuals with DBN syndrome.
Collapse
Affiliation(s)
- Elizabeth Cornforth
- MGH Institute of Health Professions, Boston, MA and Department of Physical Therapy, Spaulding Rehabilitation Hospital, Boston, MA (E.C.); and Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Ataxia Center, Cognitive Behavioral Neurology Unit, Laboratory for Neuroanatomy and Cerebellar Neurobiology, Boston, MA (J.D.S.)
| | | |
Collapse
|
3
|
Huang H, Charron TL, Fu M, Dunn M, Jones DM, Kumar P, Kulkarni A, Konopka G, Shakkottai VG. Resilience to Endoplasmic Reticulum Stress Mitigates Calcium-Dependent Membrane Hyperexcitability Underlying Late Disease Onset in SCA6. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635103. [PMID: 39975408 PMCID: PMC11838253 DOI: 10.1101/2025.01.27.635103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
An enduring puzzle in many inherited neurological disorders is the late onset of symptoms despite expression of function-impairing mutant protein early in life. We examined the basis for onset of impairment in Spinocerebellar ataxia type 6 (SCA6), a canonical late-onset neurodegenerative ataxia which results from a polyglutamine expansion in the voltage gated calcium channel, Cav2.1. Cerebellar Purkinje cell spiking abnormalities are seen concurrent with motor impairment in SCA6 mice but the basis for these changes in spiking is unknown. We identify endoplasmic reticulum (ER) calcium depletion as the cause for Purkinje cell spiking abnormalities and that the impairments in Purkinje cell spiking are unrelated to Cav2.1 ion-flux function. Further, intact inhibitory neurotransmission in the cerebellar cortex is necessary for Purkinje neurons to exhibit spiking abnormalities in SCA6 mice. Based on serial cerebellar transcriptome analysis, we define a mechanism of disease that is related to ER stress. Further, our studies support a model whereby proteotoxicity from misfolded mutant Cav2.1 is mitigated by a HSP90-dependent unfolded protein response (UPR) and that age-related breakdown of this response causes motor dysfunction and aberrant Purkinje cell spiking. Redundant pathways of the UPR mediate this resilience to ER stress. These studies elucidate a mechanism of resilience connecting aberrant proteostasis and calcium-dependent intrinsic membrane hyperexcitability to explain delayed disease onset more widely in age-dependent neurodegenerative disease. Significance Statement Advancing age is the single most important risk factor for neurodegenerative disease. Understanding how age intersects with genetic risk is therefore a critical challenge for neurodegenerative disease research. SCA6, a canonical late-onset degenerative cerebellar ataxia, results from a polyQ expansion in the voltage gated calcium channel, Cav2.1, encoded by CACNA1A . We define a mechanism of disease in SCA6 that is related to ER stress and unrelated to impaired calcium flux function of Cav2.1. Age-related decompensation of a HSP90-dependent unfolded protein response leads to disease onset. Mutant Cav2.1 misfolding as the basis for disease in SCA6 provides insight into a novel role for channelopathies to behave as proteinopathies and helps understand resilience to proteotoxicity more widely in adult-onset neurodegenerative disease.
Collapse
|
4
|
Kizeev G, Witteveen I, Balmer T. Balance Performance in Aged Mice is Dependent on Unipolar Brush Cells. CEREBELLUM (LONDON, ENGLAND) 2024; 24:16. [PMID: 39699796 DOI: 10.1007/s12311-024-01767-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
The vestibular processing regions of the cerebellum integrate vestibular information with other sensory modalities and motor signals to regulate balance, gaze stability, and spatial orientation. A class of excitatory glutamatergic interneurons known as unipolar brush cells (UBCs) are highly concentrated within the granule cell layer of these regions. UBCs receive vestibular signals directly from primary vestibular afferents and indirectly from mossy fibers. Each UBC excites numerous granule cells and could contribute to computations necessary for balance-related motor function. Prior research has implicated UBCs in motor function, but their influence on balance performance remains unclear, especially in aged mice that have age-related impairment. Here we tested whether UBCs contribute to motor coordination and balance by disrupting their activity with chemogenetics in aged and young mice. Age-related balance deficits were apparent in mice > 6 months old. Disrupting the activity of a subpopulation of UBCs caused aged mice to fall off a balance beam more frequently and altered swimming behaviors that are sensitive to vestibular dysfunction. These effects were not seen in young (7-week-old) mice. Thus, disrupting the activity of UBCs impairs mice with age-related balance issues and suggest that UBCs are essential for balance and vestibular function in aged mice.
Collapse
Affiliation(s)
- Gabrielle Kizeev
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Isabelle Witteveen
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Timothy Balmer
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
5
|
Morrison LM, Huang H, Handler HP, Fu M, Jones DM, Bushart DD, Pappas SS, Orr HT, Shakkottai VG. Increased intrinsic membrane excitability is associated with olivary hypertrophy in spinocerebellar ataxia type 1. Hum Mol Genet 2024; 33:2159-2176. [PMID: 39475127 PMCID: PMC11630738 DOI: 10.1093/hmg/ddae146] [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: 06/17/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024] Open
Abstract
One of the characteristic regions of brainstem degeneration across multiple spinocerebellar ataxias (SCAs) is the inferior olive (IO), a medullary nucleus that plays a key role in motor learning. The vulnerability of IO neurons remains a poorly-understood area of SCA pathology. In this work, we address this by evaluating IO disease in SCA1, a prototypic inherited olivopontocerebellar atrophy, using the genetically-precise SCA1 knock-in (SCA1-KI) mouse. We find that these mice exhibit olivary hypertrophy, a phenotype reminiscent of a degenerative disorder known as hypertrophic olivary degeneration (HOD). Similar to early stages of HOD, SCA1-KI IO neurons display early dendritic lengthening and later somatic expansion without frank cell loss. Though HOD is known to be caused by brainstem lesions that disrupt IO inhibitory innervation, we observe no loss of inhibitory terminals in the SCA1-KI IO. Additionally, we find that a separate mouse model of SCA1 in which mutant ATXN1 is expressed solely in cerebellar Purkinje cells shows no evidence of olivary hypertrophy. Patch-clamp recordings from brainstem slices indicate that SCA1-KI IO neurons are hyperexcitable, generating spike trains in response to membrane depolarization. Transcriptome analysis further reveals reduced medullary expression of ion channels responsible for IO neuron spike afterhyperpolarization (AHP)-a result that appears to have a functional consequence, as SCA1-KI IO neuron spikes exhibit a diminished AHP. These findings suggest that expression of mutant ATXN1 in IO neurons results in an HOD-like olivary hypertrophy, in association with increased intrinsic membrane excitability and ion channel transcriptional dysregulation.
Collapse
Affiliation(s)
- Logan M Morrison
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd. Dallas, TX 75390, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| | - Haoran Huang
- Medical Scientist Training Program, The Ohio State University, Columbus, OH 43210, United States
- College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Hillary P Handler
- Molecular Diagnostics Laboratory, University of Minnesota Fairview Medical Center, Minneapolis, MN 55455, United States
| | - Min Fu
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd. Dallas, TX 75390, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| | - Deborah M Jones
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| | - David D Bushart
- College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Samuel S Pappas
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd. Dallas, TX 75390, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| | - Harry T Orr
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, 420 Delaware Street SE, MN 55455, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, United States
| | - Vikram G Shakkottai
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd. Dallas, TX 75390, United States
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., TX 75390, United States
| |
Collapse
|
6
|
Kizeev G, Witteveen I, Balmer T. Balance performance in aged mice is dependent on unipolar brush cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617602. [PMID: 39416048 PMCID: PMC11482929 DOI: 10.1101/2024.10.10.617602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The vestibular processing regions of the cerebellum integrate vestibular information with other sensory modalities and motor signals to regulate balance, gaze stability, and spatial orientation. A class of excitatory glutamatergic interneurons known as unipolar brush cells (UBCs) are highly concentrated within the granule cell layer of these regions. UBCs receive vestibular signals directly from primary vestibular afferents and indirectly from mossy fibers. Each UBC excites numerous granule cells and could contribute to computations necessary for balance-related motor function. Prior research has implicated UBCs in motor function, but their influence on balance performance remains unclear, especially in aged mice that have age-related impairment. Here we tested whether UBCs contribute to motor coordination and balance by disrupting their activity with chemogenetics in aged and young mice. Age-related balance deficits were apparent in mice > 6 months old. Disrupting the activity of a subpopulation of UBCs caused aged mice to fall off a balance beam more frequently and altered swimming behaviors that are sensitive to vestibular dysfunction. These effects were not seen in young (7-week-old) mice. Thus, disrupting the activity of UBCs impairs mice with age-related balance issues and suggest that UBCs are essential for balance and vestibular function in aged mice.
Collapse
Affiliation(s)
- Gabrielle Kizeev
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Isabelle Witteveen
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Timothy Balmer
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| |
Collapse
|
7
|
Arnskötter F, da Silva PBG, Schouw ME, Lukasch C, Bianchini L, Sieber L, Garcia-Lopez J, Ahmad ST, Li Y, Lin H, Joshi P, Spänig L, Radoš M, Roiuk M, Sepp M, Zuckermann M, Northcott PA, Patrizi A, Kutscher LM. Loss of Elp1 in cerebellar granule cell progenitors models ataxia phenotype of Familial Dysautonomia. Neurobiol Dis 2024; 199:106600. [PMID: 38996985 DOI: 10.1016/j.nbd.2024.106600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
Familial Dysautonomia (FD) is an autosomal recessive disorder caused by a splice site mutation in the gene ELP1, which disproportionally affects neurons. While classically characterized by deficits in sensory and autonomic neurons, neuronal defects in the central nervous system have also been described. Although ELP1 expression remains high in the normal developing and adult cerebellum, its role in cerebellar development is unknown. To explore the role of Elp1 in the cerebellum, we knocked out Elp1 in cerebellar granule cell progenitors (GCPs) and examined the outcome on animal behavior and cellular composition. We found that GCP-specific conditional knockout of Elp1 (Elp1cKO) resulted in ataxia by 8 weeks of age. Cellular characterization showed that the animals had smaller cerebella with fewer granule cells. This defect was already apparent as early as 7 days after birth, when Elp1cKO animals also had fewer mitotic GCPs and shorter Purkinje dendrites. Through molecular characterization, we found that loss of Elp1 was associated with an increase in apoptotic cell death and cell stress pathways in GCPs. Our study demonstrates the importance of ELP1 in the developing cerebellum, and suggests that loss of Elp1 in the GC lineage may also play a role in the progressive ataxia phenotypes of FD patients.
Collapse
Affiliation(s)
- Frederik Arnskötter
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Patricia Benites Goncalves da Silva
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany
| | - Mackenna E Schouw
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany
| | - Chiara Lukasch
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany
| | - Luca Bianchini
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Laura Sieber
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany
| | - Jesus Garcia-Lopez
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA; Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN, USA; Department of In vivo Pharmacology-Immunology, Tempest Therapeutics, Brisbane, CA, USA
| | - Shiekh Tanveer Ahmad
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA; Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yiran Li
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA; Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hong Lin
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA; Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Piyush Joshi
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany
| | - Lisa Spänig
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Magdalena Radoš
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany
| | - Mykola Roiuk
- Signal Transduction in Cancer and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mari Sepp
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Marc Zuckermann
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany; Division of Pediatric Neuro-Oncology, Preclinical Modeling Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paul A Northcott
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA; Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Annarita Patrizi
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena M Kutscher
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, A partnership between DKFZ and Heidelberg University Hospital, Germany.
| |
Collapse
|
8
|
Li ZH, Li B, Zhang XY, Zhu JN. Neuropeptides and Their Roles in the Cerebellum. Int J Mol Sci 2024; 25:2332. [PMID: 38397008 PMCID: PMC10889816 DOI: 10.3390/ijms25042332] [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: 01/10/2024] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Although more than 30 different types of neuropeptides have been identified in various cell types and circuits of the cerebellum, their unique functions in the cerebellum remain poorly understood. Given the nature of their diffuse distribution, peptidergic systems are generally assumed to exert a modulatory effect on the cerebellum via adaptively tuning neuronal excitability, synaptic transmission, and synaptic plasticity within cerebellar circuits. Moreover, cerebellar neuropeptides have also been revealed to be involved in the neurogenetic and developmental regulation of the developing cerebellum, including survival, migration, differentiation, and maturation of the Purkinje cells and granule cells in the cerebellar cortex. On the other hand, cerebellar neuropeptides hold a critical position in the pathophysiology and pathogenesis of many cerebellar-related motor and psychiatric disorders, such as cerebellar ataxias and autism. Over the past two decades, a growing body of evidence has indicated neuropeptides as potential therapeutic targets to ameliorate these diseases effectively. Therefore, this review focuses on eight cerebellar neuropeptides that have attracted more attention in recent years and have significant potential for clinical application associated with neurodegenerative and/or neuropsychiatric disorders, including brain-derived neurotrophic factor, corticotropin-releasing factor, angiotensin II, neuropeptide Y, orexin, thyrotropin-releasing hormone, oxytocin, and secretin, which may provide novel insights and a framework for our understanding of cerebellar-related disorders and have implications for novel treatments targeting neuropeptide systems.
Collapse
Affiliation(s)
- Zi-Hao Li
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing 210023, China; (Z.-H.L.); (J.-N.Z.)
| | - Bin Li
- Women and Children’s Medical Research Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiao-Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing 210023, China; (Z.-H.L.); (J.-N.Z.)
- Institute for Brain Sciences, Nanjing University, Nanjing 210023, China
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing 210023, China; (Z.-H.L.); (J.-N.Z.)
- Institute for Brain Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
9
|
Pilotto F, Del Bondio A, Puccio H. Hereditary Ataxias: From Bench to Clinic, Where Do We Stand? Cells 2024; 13:319. [PMID: 38391932 PMCID: PMC10886822 DOI: 10.3390/cells13040319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
Cerebellar ataxias are a wide heterogeneous group of movement disorders. Within this broad umbrella of diseases, there are both genetics and sporadic forms. The clinical presentation of these conditions can exhibit a diverse range of symptoms across different age groups, spanning from pure cerebellar manifestations to sensory ataxia and multisystemic diseases. Over the last few decades, advancements in our understanding of genetics and molecular pathophysiology related to both dominant and recessive ataxias have propelled the field forward, paving the way for innovative therapeutic strategies aimed at preventing and arresting the progression of these diseases. Nevertheless, the rarity of certain forms of ataxia continues to pose challenges, leading to limited insights into the etiology of the disease and the identification of target pathways. Additionally, the lack of suitable models hampers efforts to comprehensively understand the molecular foundations of disease's pathophysiology and test novel therapeutic interventions. In the following review, we describe the epidemiology, symptomatology, and pathological progression of hereditary ataxia, including both the prevalent and less common forms of these diseases. Furthermore, we illustrate the diverse molecular pathways and therapeutic approaches currently undergoing investigation in both pre-clinical studies and clinical trials. Finally, we address the existing and anticipated challenges within this field, encompassing both basic research and clinical endeavors.
Collapse
Affiliation(s)
| | | | - Hélène Puccio
- Institut Neuromyogène, Pathophysiology and Genetics of Neuron and Muscle, Inserm U1315, CNRS-Université Claude Bernard Lyon 1 UMR5261, 69008 Lyon, France
| |
Collapse
|
10
|
Tejwani L, Ravindra NG, Lee C, Cheng Y, Nguyen B, Luttik K, Ni L, Zhang S, Morrison LM, Gionco J, Xiang Y, Yoon J, Ro H, Haidery F, Grijalva RM, Bae E, Kim K, Martuscello RT, Orr HT, Zoghbi HY, McLoughlin HS, Ranum LPW, Shakkottai VG, Faust PL, Wang S, van Dijk D, Lim J. Longitudinal single-cell transcriptional dynamics throughout neurodegeneration in SCA1. Neuron 2024; 112:362-383.e15. [PMID: 38016472 PMCID: PMC10922326 DOI: 10.1016/j.neuron.2023.10.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 09/10/2023] [Accepted: 10/27/2023] [Indexed: 11/30/2023]
Abstract
Neurodegeneration is a protracted process involving progressive changes in myriad cell types that ultimately results in the death of vulnerable neuronal populations. To dissect how individual cell types within a heterogeneous tissue contribute to the pathogenesis and progression of a neurodegenerative disorder, we performed longitudinal single-nucleus RNA sequencing of mouse and human spinocerebellar ataxia type 1 (SCA1) cerebellar tissue, establishing continuous dynamic trajectories of each cell population. Importantly, we defined the precise transcriptional changes that precede loss of Purkinje cells and, for the first time, identified robust early transcriptional dysregulation in unipolar brush cells and oligodendroglia. Finally, we applied a deep learning method to predict disease state accurately and identified specific features that enable accurate distinction of wild-type and SCA1 cells. Together, this work reveals new roles for diverse cerebellar cell types in SCA1 and provides a generalizable analysis framework for studying neurodegeneration.
Collapse
Affiliation(s)
- Leon Tejwani
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Neal G Ravindra
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Computer Science, Yale University, New Haven, CT 06510, USA
| | - Changwoo Lee
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yubao Cheng
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Billy Nguyen
- University of California, San Francisco School of Medicine, San Francisco, CA 94143, USA
| | - Kimberly Luttik
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Luhan Ni
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Shupei Zhang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Logan M Morrison
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - John Gionco
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY 10032, USA
| | - Yangfei Xiang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Hannah Ro
- Yale College, New Haven, CT 06510, USA
| | | | - Rosalie M Grijalva
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Kristen Kim
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA
| | - Regina T Martuscello
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY 10032, USA
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Huda Y Zoghbi
- 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; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hayley S McLoughlin
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Laura P W Ranum
- Department of Molecular Genetics and Microbiology, Center for Neurogenetics, College of Medicine, Genetics Institute, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Vikram G Shakkottai
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Phyllis L Faust
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY 10032, USA
| | - Siyuan Wang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA.
| | - David van Dijk
- Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Computer Science, Yale University, New Haven, CT 06510, USA.
| | - Janghoo Lim
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510, USA; Wu Tsai Institute, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
11
|
Lalonde R, Hernandez M, Strazielle C. BDNF and Cerebellar Ataxia. Curr Drug Res Rev 2024; 16:300-307. [PMID: 37609676 DOI: 10.2174/2589977515666230811093021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 08/24/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) has been proposed as a treatment for neurodegeneration, including diseases of the cerebellum, where BDNF levels or those of its main receptor, TrkB, are often diminished relative to controls, thereby serving as replacement therapy. Experimental evidence indicates that BDNF signaling countered cerebellar degeneration, sensorimotor deficits, or both, in transgenic ATXN1 mice mutated for ataxin-1, Cacna1a knock-in mice mutated for ataxin-6, mice injected with lentivectors encoding RNA sequences against human FXN into the cerebellar cortex, Kcnj6Wv (Weaver) mutant mice with granule cell degeneration, and rats with olivocerebellar transaction, similar to a BDNF-overexpressing transgenic line interbred with Cacng2stg mutant mice. In this regard, this study discusses whether BDNF is effective in cerebellar pathologies where BDNF levels are normal and whether it is effective in cases with combined cerebellar and basal ganglia damage.
Collapse
Affiliation(s)
- Robert Lalonde
- Université de Lorraine, Laboratoire Stress, Immunité, Pathogènes EA 7300, Campus Santé, 9 avenue de la Forêt de Haye, 54500 Vandoeuvre-les-Nancy, France
| | - Magali Hernandez
- Université de Lorraine, Laboratoire Stress, Immunité, Pathogènes EA 7300, Campus Santé, 9 avenue de la Forêt de Haye, 54500 Vandoeuvre-les-Nancy, France
- CHRU Nancy, allée du Morvan, 54500 Vandoeuvre-les-Nancy, France
| | - Catherine Strazielle
- Université de Lorraine, Laboratoire Stress, Immunité, Pathogènes EA 7300, Campus Santé, 9 avenue de la Forêt de Haye, 54500 Vandoeuvre-les-Nancy, France
- CHRU Nancy, allée du Morvan, 54500 Vandoeuvre-les-Nancy, France
| |
Collapse
|
12
|
Pellerin D, Danzi MC, Renaud M, Houlden H, Synofzik M, Zuchner S, Brais B. Spinocerebellar ataxia 27B: A novel, frequent and potentially treatable ataxia. Clin Transl Med 2024; 14:e1504. [PMID: 38279833 PMCID: PMC10819088 DOI: 10.1002/ctm2.1504] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/19/2023] [Accepted: 11/24/2023] [Indexed: 01/29/2024] Open
Abstract
Hereditary ataxias, especially when presenting sporadically in adulthood, present a particular diagnostic challenge owing to their great clinical and genetic heterogeneity. Currently, up to 75% of such patients remain without a genetic diagnosis. In an era of emerging disease-modifying gene-stratified therapies, the identification of causative alleles has become increasingly important. Over the past few years, the implementation of advanced bioinformatics tools and long-read sequencing has allowed the identification of a number of novel repeat expansion disorders, such as the recently described spinocerebellar ataxia 27B (SCA27B) caused by a (GAA)•(TTC) repeat expansion in intron 1 of the fibroblast growth factor 14 (FGF14) gene. SCA27B is rapidly gaining recognition as one of the most common forms of adult-onset hereditary ataxia, with several studies showing that it accounts for a substantial number (9-61%) of previously undiagnosed cases from different cohorts. First natural history studies and multiple reports have already outlined the progression and core phenotype of this novel disease, which consists of a late-onset slowly progressive pan-cerebellar syndrome that is frequently associated with cerebellar oculomotor signs, such as downbeat nystagmus, and episodic symptoms. Furthermore, preliminary studies in patients with SCA27B have shown promising symptomatic benefits of 4-aminopyridine, an already marketed drug. This review describes the current knowledge of the genetic and molecular basis, epidemiology, clinical features and prospective treatment strategies in SCA27B.
Collapse
Affiliation(s)
- David Pellerin
- Department of Neurology and Neurosurgery, Montreal Neurological Hospital and InstituteMcGill UniversityMontrealQuebecCanada
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and NeurosurgeryUniversity College LondonLondonUK
| | - Matt C. Danzi
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Mathilde Renaud
- INSERM‐U1256 NGEREUniversité de LorraineNancyFrance
- Service de Neurologie, CHRU de NancyNancyFrance
- Service de Génétique Clinique, CHRU de NancyNancyFrance
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and NeurosurgeryUniversity College LondonLondonUK
| | - Matthis Synofzik
- Division of Translational Genomics of Neurodegenerative DiseasesHertie‐Institute for Clinical Brain Research and Center of Neurology, University of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Stephan Zuchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human GenomicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Bernard Brais
- Department of Neurology and Neurosurgery, Montreal Neurological Hospital and InstituteMcGill UniversityMontrealQuebecCanada
- Department of Human GeneticsMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
13
|
Kumar M, Tyagi N, Faruq M. The molecular mechanisms of spinocerebellar ataxias for DNA repeat expansion in disease. Emerg Top Life Sci 2023; 7:289-312. [PMID: 37668011 DOI: 10.1042/etls20230013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023]
Abstract
Spinocerebellar ataxias (SCAs) are a heterogenous group of neurodegenerative disorders which commonly inherited in an autosomal dominant manner. They cause muscle incoordination due to degeneration of the cerebellum and other parts of nervous system. Out of all the characterized (>50) SCAs, 14 SCAs are caused due to microsatellite repeat expansion mutations. Repeat expansions can result in toxic protein gain-of-function, protein loss-of-function, and/or RNA gain-of-function effects. The location and the nature of mutation modulate the underlying disease pathophysiology resulting in varying disease manifestations. Potential toxic effects of these mutations likely affect key major cellular processes such as transcriptional regulation, mitochondrial functioning, ion channel dysfunction and synaptic transmission. Involvement of several common pathways suggests interlinked function of genes implicated in the disease pathogenesis. A better understanding of the shared and distinct molecular pathogenic mechanisms in these diseases is required to develop targeted therapeutic tools and interventions for disease management. The prime focus of this review is to elaborate on how expanded 'CAG' repeats contribute to the common modes of neurotoxicity and their possible therapeutic targets in management of such devastating disorders.
Collapse
Affiliation(s)
- Manish Kumar
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| | - Nishu Tyagi
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| | - Mohammed Faruq
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| |
Collapse
|
14
|
Morrison LM, Huang H, Handler HP, Fu M, Bushart DD, Pappas SS, Orr HT, Shakkottai VG. Increased intrinsic membrane excitability is associated with hypertrophic olivary degeneration in spinocerebellar ataxia type 1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563657. [PMID: 37961407 PMCID: PMC10634770 DOI: 10.1101/2023.10.23.563657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
One of the characteristic areas of brainstem degeneration across multiple spinocerebellar ataxias (SCAs) is the inferior olive (IO), a medullary nucleus that plays a key role in motor learning. In addition to its vulnerability in SCAs, the IO is also susceptible to a distinct pathology known as hypertrophic olivary degeneration (HOD). Clinically, HOD has been exclusively observed after lesions in the brainstem disrupt inhibitory afferents to the IO. Here, for the first time, we describe HOD in another context: spinocerebellar ataxia type 1 (SCA1). Using the genetically-precise SCA1 knock-in mouse model (SCA1-KI; both sexes used), we assessed SCA1-associated changes in IO neuron structure and function. Concurrent with degeneration, we found that SCA1-KI IO neurons are hypertrophic, exhibiting early dendrite lengthening and later somatic expansion. Unlike in previous descriptions of HOD, we observed no clear loss of IO inhibitory innervation; nevertheless, patch-clamp recordings from brainstem slices reveal that SCA1-KI IO neurons are hyperexcitable. Rather than synaptic disinhibition, we identify increases in intrinsic membrane excitability as the more likely mechanism underlying this novel SCA1 phenotype. Specifically, transcriptome analysis indicates that SCA1-KI IO hyperexcitability is associated with a reduced medullary expression of ion channels responsible for spike afterhyperpolarization (AHP) in IO neurons - a result that has a functional consequence, as SCA1-KI IO neuron spikes exhibit a diminished AHP. These results reveal membrane excitability as a potential link between disparate causes of IO degeneration, suggesting that HOD can result from any cause, intrinsic or extrinsic, that increases excitability of the IO neuron membrane.
Collapse
Affiliation(s)
- Logan M. Morrison
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Haoran Huang
- Medical Scientist Training Program, The Ohio State University, Columbus, OH 43210 USA
- College of Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Hillary P. Handler
- Molecular Diagnostics Laboratory, University of Minnesota Fairview Medical Center, Minneapolis, MN 55455, USA
| | - Min Fu
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David D. Bushart
- College of Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Samuel S. Pappas
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Harry T. Orr
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vikram G. Shakkottai
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
15
|
Nanclares C, Noriega-Prieto JA, Labrada-Moncada FE, Cvetanovic M, Araque A, Kofuji P. Altered calcium signaling in Bergmann glia contributes to spinocerebellar ataxia type-1 in a mouse model of SCA1. Neurobiol Dis 2023; 187:106318. [PMID: 37802154 PMCID: PMC10624966 DOI: 10.1016/j.nbd.2023.106318] [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: 08/15/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by an abnormal expansion of glutamine (Q) encoding CAG repeats in the ATAXIN1 (ATXN1) gene and characterized by progressive cerebellar ataxia, dysarthria, and eventual deterioration of bulbar functions. SCA1 shows severe degeneration of cerebellar Purkinje cells (PCs) and activation of Bergmann glia (BG), a type of cerebellar astroglia closely associated with PCs. Combining electrophysiological recordings, calcium imaging techniques, and chemogenetic approaches, we have investigated the electrical intrinsic and synaptic properties of PCs and the physiological properties of BG in SCA1 mouse model expressing mutant ATXN1 only in PCs. PCs of SCA1 mice displayed lower spontaneous firing rate and larger slow afterhyperpolarization currents (sIAHP) than wildtype mice, whereas the properties of the synaptic inputs were unaffected. BG of SCA1 mice showed higher calcium hyperactivity and gliotransmission, manifested by higher frequency of NMDAR-mediated slow inward currents (SICs) in PC. Preventing the BG calcium hyperexcitability of SCA1 mice by loading BG with the calcium chelator BAPTA restored sIAHP and spontaneous firing rate of PCs to similar levels of wildtype mice. Moreover, mimicking the BG hyperactivity by activating BG expressing Gq-DREADDs in wildtype mice reproduced the SCA1 pathological phenotype of PCs, i.e., enhancement of sIAHP and decrease of spontaneous firing rate. These results indicate that the intrinsic electrical properties of PCs, but not their synaptic properties, were altered in SCA1 mice and that these alterations were associated with the hyperexcitability of BG. Moreover, preventing BG hyperexcitability in SCA1 mice and promoting BG hyperexcitability in wildtype mice prevented and mimicked, respectively, the pathological electrophysiological phenotype of PCs. Therefore, BG plays a relevant role in the dysfunction of the electrical intrinsic properties of PCs in SCA1 mice, suggesting that they may serve as potential targets for therapeutic approaches to treat the spinocerebellar ataxia type 1.
Collapse
Affiliation(s)
- Carmen Nanclares
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
16
|
Bremova-Ertl T, Hofmann J, Stucki J, Vossenkaul A, Gautschi M. Inborn Errors of Metabolism with Ataxia: Current and Future Treatment Options. Cells 2023; 12:2314. [PMID: 37759536 PMCID: PMC10527548 DOI: 10.3390/cells12182314] [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: 08/15/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
A number of hereditary ataxias are caused by inborn errors of metabolism (IEM), most of which are highly heterogeneous in their clinical presentation. Prompt diagnosis is important because disease-specific therapies may be available. In this review, we offer a comprehensive overview of metabolic ataxias summarized by disease, highlighting novel clinical trials and emerging therapies with a particular emphasis on first-in-human gene therapies. We present disease-specific treatments if they exist and review the current evidence for symptomatic treatments of these highly heterogeneous diseases (where cerebellar ataxia is part of their phenotype) that aim to improve the disease burden and enhance quality of life. In general, a multimodal and holistic approach to the treatment of cerebellar ataxia, irrespective of etiology, is necessary to offer the best medical care. Physical therapy and speech and occupational therapy are obligatory. Genetic counseling is essential for making informed decisions about family planning.
Collapse
Affiliation(s)
- Tatiana Bremova-Ertl
- Department of Neurology, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland; (J.H.); (J.S.)
- Center for Rare Diseases, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland
| | - Jan Hofmann
- Department of Neurology, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland; (J.H.); (J.S.)
| | - Janine Stucki
- Department of Neurology, University Hospital Bern (Inselspital) and University of Bern, 3010 Bern, Switzerland; (J.H.); (J.S.)
| | - Anja Vossenkaul
- Division of Pediatric Endocrinology, Diabetes and Metabolism, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (A.V.); (M.G.)
| | - Matthias Gautschi
- Division of Pediatric Endocrinology, Diabetes and Metabolism, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (A.V.); (M.G.)
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
17
|
Buijsen RAM, Hu M, Sáez-González M, Notopoulou S, Mina E, Koning W, Gardiner SL, van der Graaf LM, Daoutsali E, Pepers BA, Mei H, van Dis V, Frimat JP, van den Maagdenberg AMJM, Petrakis S, van Roon-Mom WMC. Spinocerebellar Ataxia Type 1 Characteristics in Patient-Derived Fibroblast and iPSC-Derived Neuronal Cultures. Mov Disord 2023; 38:1428-1442. [PMID: 37278528 DOI: 10.1002/mds.29446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/23/2023] [Accepted: 04/20/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by a polyglutamine expansion in the ataxin-1 protein resulting in neuropathology including mutant ataxin-1 protein aggregation, aberrant neurodevelopment, and mitochondrial dysfunction. OBJECTIVES Identify SCA1-relevant phenotypes in patient-specific fibroblasts and SCA1 induced pluripotent stem cells (iPSCs) neuronal cultures. METHODS SCA1 iPSCs were generated and differentiated into neuronal cultures. Protein aggregation and neuronal morphology were evaluated using fluorescent microscopy. Mitochondrial respiration was measured using the Seahorse Analyzer. The multi-electrode array (MEA) was used to identify network activity. Finally, gene expression changes were studied using RNA-seq to identify disease-specific mechanisms. RESULTS Bioenergetics deficits in patient-derived fibroblasts and SCA1 neuronal cultures showed altered oxygen consumption rate, suggesting involvement of mitochondrial dysfunction in SCA1. In SCA1 hiPSC-derived neuronal cells, nuclear and cytoplasmic aggregates were identified similar in localization as aggregates in SCA1 postmortem brain tissue. SCA1 hiPSC-derived neuronal cells showed reduced dendrite length and number of branching points while MEA recordings identified delayed development in network activity in SCA1 hiPSC-derived neuronal cells. Transcriptome analysis identified 1050 differentially expressed genes in SCA1 hiPSC-derived neuronal cells associated with synapse organization and neuron projection guidance, where a subgroup of 151 genes was highly associated with SCA1 phenotypes and linked to SCA1 relevant signaling pathways. CONCLUSIONS Patient-derived cells recapitulate key pathological features of SCA1 pathogenesis providing a valuable tool for the identification of novel disease-specific processes. This model can be used for high throughput screenings to identify compounds, which may prevent or rescue neurodegeneration in this devastating disease. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ronald A M Buijsen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Michel Hu
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Maria Sáez-González
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sofia Notopoulou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Eleni Mina
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Winette Koning
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sarah L Gardiner
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Linda M van der Graaf
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Elena Daoutsali
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Barry A Pepers
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Hailiang Mei
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Vera van Dis
- Department of Pathology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Pathology, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Jean-Philippe Frimat
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Spyros Petrakis
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Willeke M C van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| |
Collapse
|
18
|
Kapfhammer JP, Shimobayashi E. Viewpoint: spinocerebellar ataxias as diseases of Purkinje cell dysfunction rather than Purkinje cell loss. Front Mol Neurosci 2023; 16:1182431. [PMID: 37426070 PMCID: PMC10323145 DOI: 10.3389/fnmol.2023.1182431] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/22/2023] [Indexed: 07/11/2023] Open
Abstract
Spinocerebellar ataxias (SCAs) are a group of hereditary neurodegenerative diseases mostly affecting cerebellar Purkinje cells caused by a wide variety of different mutations. One subtype, SCA14, is caused by mutations of Protein Kinase C gamma (PKCγ), the dominant PKC isoform present in Purkinje cells. Mutations in the pathway in which PKCγ is active, i.e., in the regulation of calcium levels and calcium signaling in Purkinje cells, are the cause of several other variants of SCA. In SCA14, many of the observed mutations in the PKCγ gene were shown to increase the basal activity of PKCγ, raising the possibility that increased activity of PKCγ might be the cause of most forms of SCA14 and might also be involved in the pathogenesis of SCA in related subtypes. In this viewpoint and review article we will discuss the evidence for and against such a major role of PKCγ basal activity and will suggest a hypothesis of how PKCγ activity and the calcium signaling pathway may be involved in the pathogenesis of SCAs despite the different and sometimes opposing effects of mutations affecting these pathways. We will then widen the scope and propose a concept of SCA pathogenesis which is not primarily driven by cell death and loss of Purkinje cells but rather by dysfunction of Purkinje cells which are still present and alive in the cerebellum.
Collapse
|
19
|
Cundari M, Vestberg S, Gustafsson P, Gorcenco S, Rasmussen A. Neurocognitive and cerebellar function in ADHD, autism and spinocerebellar ataxia. Front Syst Neurosci 2023; 17:1168666. [PMID: 37415926 PMCID: PMC10321758 DOI: 10.3389/fnsys.2023.1168666] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023] Open
Abstract
The cerebellum plays a major role in balance, motor control and sensorimotor integration, but also in cognition, language, and emotional regulation. Several neuropsychiatric disorders such as attention deficit-hyperactivity disorder (ADHD), autism spectrum disorder (ASD), as well as neurological diseases such as spinocerebellar ataxia type 3 (SCA3) are associated with differences in cerebellar function. Morphological abnormalities in different cerebellar subregions produce distinct behavioral symptoms related to the functional disruption of specific cerebro-cerebellar circuits. The specific contribution of the cerebellum to typical development may therefore involve the optimization of the structure and function of cerebro-cerebellar circuits underlying skill acquisition in multiple domains. Here, we review cerebellar structural and functional differences between healthy and patients with ADHD, ASD, and SCA3, and explore how disruption of cerebellar networks affects the neurocognitive functions in these conditions. We discuss how cerebellar computations contribute to performance on cognitive and motor tasks and how cerebellar signals are interfaced with signals from other brain regions during normal and dysfunctional behavior. We conclude that the cerebellum plays a role in many cognitive functions. Still, more clinical studies with the support of neuroimaging are needed to clarify the cerebellum's role in normal and dysfunctional behavior and cognitive functioning.
Collapse
Affiliation(s)
- Maurizio Cundari
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
- Unit of Neuropsychiatry, Hospital of Helsingborg, Helsingborg, Sweden
- Unit of Neurology, Hospital of Helsingborg, Helsingborg, Sweden
| | - Susanna Vestberg
- Department of Psychology, Faculty of Social Science, Lund University, Lund, Sweden
| | - Peik Gustafsson
- Child and Adolescent Psychiatry, Department of Clinical Sciences Lund, Medical Faculty, Lund University, Lund, Sweden
| | - Sorina Gorcenco
- Department for Clinical Sciences Lund, Neurology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Anders Rasmussen
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
20
|
Huang H, Shakkottai VG. Targeting Ion Channels and Purkinje Neuron Intrinsic Membrane Excitability as a Therapeutic Strategy for Cerebellar Ataxia. Life (Basel) 2023; 13:1350. [PMID: 37374132 DOI: 10.3390/life13061350] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
In degenerative neurological disorders such as Parkinson's disease, a convergence of widely varying insults results in a loss of dopaminergic neurons and, thus, the motor symptoms of the disease. Dopamine replacement therapy with agents such as levodopa is a mainstay of therapy. Cerebellar ataxias, a heterogeneous group of currently untreatable conditions, have not been identified to have a shared physiology that is a target of therapy. In this review, we propose that perturbations in cerebellar Purkinje neuron intrinsic membrane excitability, a result of ion channel dysregulation, is a common pathophysiologic mechanism that drives motor impairment and vulnerability to degeneration in cerebellar ataxias of widely differing genetic etiologies. We further propose that treatments aimed at restoring Purkinje neuron intrinsic membrane excitability have the potential to be a shared therapy in cerebellar ataxia akin to levodopa for Parkinson's disease.
Collapse
Affiliation(s)
- Haoran Huang
- Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Vikram G Shakkottai
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
21
|
Osório C, White JJ, Lu H, Beekhof GC, Fiocchi FR, Andriessen CA, Dijkhuizen S, Post L, Schonewille M. Pre-ataxic loss of intrinsic plasticity and motor learning in a mouse model of SCA1. Brain 2023; 146:2332-2345. [PMID: 36352508 PMCID: PMC10232256 DOI: 10.1093/brain/awac422] [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: 04/16/2022] [Revised: 10/04/2022] [Accepted: 10/24/2022] [Indexed: 12/29/2023] Open
Abstract
Spinocerebellar ataxias are neurodegenerative diseases, the hallmark symptom of which is the development of ataxia due to cerebellar dysfunction. Purkinje cells, the principal neurons of the cerebellar cortex, are the main cells affected in these disorders, but the sequence of pathological events leading to their dysfunction is poorly understood. Understanding the origins of Purkinje cells dysfunction before it manifests is imperative to interpret the functional and behavioural consequences of cerebellar-related disorders, providing an optimal timeline for therapeutic interventions. Here, we report the cascade of events leading to Purkinje cells dysfunction before the onset of ataxia in a mouse model of spinocerebellar ataxia 1 (SCA1). Spatiotemporal characterization of the ATXN1[82Q] SCA1 mouse model revealed high levels of the mutant ATXN1[82Q] weeks before the onset of ataxia. The expression of the toxic protein first caused a reduction of Purkinje cells intrinsic excitability, which was followed by atrophy of Purkinje cells dendrite arborization and aberrant glutamatergic signalling, finally leading to disruption of Purkinje cells innervation of climbing fibres and loss of intrinsic plasticity of Purkinje cells. Functionally, we found that deficits in eyeblink conditioning, a form of cerebellum-dependent motor learning, precede the onset of ataxia, matching the timeline of climbing fibre degeneration and reduced intrinsic plasticity. Together, our results suggest that abnormal synaptic signalling and intrinsic plasticity during the pre-ataxia stage of spinocerebellar ataxias underlie an aberrant cerebellar circuitry that anticipates the full extent of the disease severity. Furthermore, our work indicates the potential for eyeblink conditioning to be used as a sensitive tool to detect early cerebellar dysfunction as a sign of future disease.
Collapse
Affiliation(s)
- Catarina Osório
- Department of Neuroscience, Erasmus Medical Center, Rotterdam 3015CN, The Netherlands
| | - Joshua J White
- Department of Neuroscience, Erasmus Medical Center, Rotterdam 3015CN, The Netherlands
| | - Heiling Lu
- Department of Neuroscience, Erasmus Medical Center, Rotterdam 3015CN, The Netherlands
| | - Gerrit C Beekhof
- Department of Neuroscience, Erasmus Medical Center, Rotterdam 3015CN, The Netherlands
| | | | | | - Stephanie Dijkhuizen
- Department of Neuroscience, Erasmus Medical Center, Rotterdam 3015CN, The Netherlands
| | - Laura Post
- Department of Neuroscience, Erasmus Medical Center, Rotterdam 3015CN, The Netherlands
| | - Martijn Schonewille
- Department of Neuroscience, Erasmus Medical Center, Rotterdam 3015CN, The Netherlands
| |
Collapse
|
22
|
Ma T, Feng L, Wei S, Wang Y, Li G, Lu Y, Zhang Y, Chu Y, Wang W, Zhang H. Antisense oligonucleotides targeting basal forebrain ATXN2 enhances spatial memory and ameliorates sleep deprivation-induced fear memory impairment in mice. Brain Behav 2023; 13:e3013. [PMID: 37072935 PMCID: PMC10275523 DOI: 10.1002/brb3.3013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/04/2022] [Accepted: 12/24/2022] [Indexed: 04/20/2023] Open
Abstract
INTRODUCTION Regulation of brain-derived neurotrophic factor (BDNF) in the basal forebrain ameliorates sleep deprivation-induced fear memory impairments in rodents. Antisense oligonucleotides (ASOs) targeting ATXN2 was a potential therapy for spinocerebellar ataxia, whose pathogenic mechanism associates with reduced BDNF expression. We tested the hypothesis that ASO7 targeting ATXN2 could affect BDNF levels in mouse basal forebrain and ameliorate sleep deprivation-induced fear memory impairments. METHODS Adult male C57BL/6 mice were used to evaluate the effects of ASO7 targeting ATXN2 microinjected into the bilateral basal forebrain (1 μg, 0.5 μL, each side) on spatial memory, fear memory and sleep deprivation-induced fear memory impairments. Spatial memory and fear memory were detected by the Morris water maze and step-down inhibitory avoidance test, respectively. Immunohistochemistry, RT-PCR, and Western blot were used to evaluate the changes of levels of BDNF, ATXN2, and postsynaptic density 95 (PSD95) protein as well as ATXN2 mRNA. The morphological changes in neurons in the hippocampal CA1 region were detected by HE staining and Nissl staining. RESULTS ASO7 targeting ATXN2 microinjected into the basal forebrain could suppress ATXN2 mRNA and protein expression for more than 1 month and enhance spatial memory but not fear memory in mice. BDNF mRNA and protein expression in basal forebrain and hippocampus was increased by ASO7. Moreover, PSD95 expression and synapse formation were increased in the hippocampus. Furthermore, ASO7 microinjected into the basal forebrain increased BDNF and PSD95 protein expression in the basal forebrain of sleep-deprived mice and counteracted sleep deprivation-induced fear memory impairments. CONCLUSION ASOs targeting ATXN2 may provide effective interventions for sleep deprivation-induced cognitive impairments.
Collapse
Affiliation(s)
- Tao Ma
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Long Feng
- Department of AnesthesiologyPLA General Hospital of Hainan HospitalHainanChina
| | - Shi‐Nan Wei
- PLA Rocket Force Characteristic Medical Center, Postgraduate Training Base of Jinzhou Medical UniversityBeijingChina
| | - Ying‐Ying Wang
- Department of AnesthesiologyBeijing Ditan Hospital, Capital Medical UniversityBeijingChina
| | - Guan‐Hua Li
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Yan Lu
- Department of NeurologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Ying‐Xin Zhang
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Yang Chu
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Wei Wang
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| | - Hao Zhang
- Department of AnesthesiologyPLA Rocket Force Characteristic Medical CenterBeijingChina
| |
Collapse
|
23
|
Lee JH, Khan MM, Stark AP, Seo S, Norton A, Yao Z, Chen CH, Regehr WG. Cerebellar granule cell signaling is indispensable for normal motor performance. Cell Rep 2023; 42:112429. [PMID: 37141091 PMCID: PMC10258556 DOI: 10.1016/j.celrep.2023.112429] [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: 08/17/2022] [Revised: 02/06/2023] [Accepted: 04/07/2023] [Indexed: 05/05/2023] Open
Abstract
Within the cerebellar cortex, mossy fibers (MFs) excite granule cells (GCs) that excite Purkinje cells (PCs), which provide outputs to the deep cerebellar nuclei (DCNs). It is well established that PC disruption produces motor deficits such as ataxia. This could arise from either decreases in ongoing PC-DCN inhibition, increases in the variability of PC firing, or disruption of the flow of MF-evoked signals. Remarkably, it is not known whether GCs are essential for normal motor function. Here we address this issue by selectively eliminating calcium channels that mediate transmission (CaV2.1, CaV2.2, and CaV2.3) in a combinatorial manner. We observe profound motor deficits but only when all CaV2 channels are eliminated. In these mice, the baseline rate and variability of PC firing are unaltered, and locomotion-dependent increases in PC firing are eliminated. We conclude that GCs are indispensable for normal motor performance and that disruption of MF-induced signals impairs motor performance.
Collapse
Affiliation(s)
- Joon-Hyuk Lee
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Mehak M Khan
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Amanda P Stark
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Soobin Seo
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Aliya Norton
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Zhiyi Yao
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher H Chen
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Kerkhof LMC, van de Warrenburg BPC, van Roon-Mom WMC, Buijsen RAM. Therapeutic Strategies for Spinocerebellar Ataxia Type 1. Biomolecules 2023; 13:biom13050788. [PMID: 37238658 DOI: 10.3390/biom13050788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is an autosomal dominant neurodegenerative disorder that affects one or two individuals per 100,000. The disease is caused by an extended CAG repeat in exon 8 of the ATXN1 gene and is characterized mostly by a profound loss of cerebellar Purkinje cells, leading to disturbances in coordination, balance, and gait. At present, no curative treatment is available for SCA1. However, increasing knowledge on the cellular and molecular mechanisms of SCA1 has led the way towards several therapeutic strategies that can potentially slow disease progression. SCA1 therapeutics can be classified as genetic, pharmacological, and cell replacement therapies. These different therapeutic strategies target either the (mutant) ATXN1 RNA or the ataxin-1 protein, pathways that play an important role in downstream SCA1 disease mechanisms or which help restore cells that are lost due to SCA1 pathology. In this review, we will provide a summary of the different therapeutic strategies that are currently being investigated for SCA1.
Collapse
Affiliation(s)
- Laurie M C Kerkhof
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Dutch Center for RNA Therapeutics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Bart P C van de Warrenburg
- Department of Neurology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Willeke M C van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Dutch Center for RNA Therapeutics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Ronald A M Buijsen
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
25
|
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
|
26
|
Cook AA, Jayabal S, Sheng J, Fields E, Leung TCS, Quilez S, McNicholas E, Lau L, Huang S, Watt AJ. Activation of TrkB-Akt signaling rescues deficits in a mouse model of SCA6. SCIENCE ADVANCES 2022; 8:eabh3260. [PMID: 36112675 PMCID: PMC9481119 DOI: 10.1126/sciadv.abh3260] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/29/2022] [Indexed: 06/01/2023]
Abstract
Spinocerebellar ataxia type 6 (SCA6) is a neurodegenerative disease resulting in motor coordination deficits and cerebellar pathology. Expression of brain-derived neurotrophic factor (BDNF) is reduced in postmortem tissue from SCA6 patients. Here, we show that levels of cerebellar BDNF and its receptor, tropomyosin receptor kinase B (TrkB), are reduced at an early disease stage in a mouse model of SCA6 (SCA684Q/84Q). One month of exercise elevated cerebellar BDNF expression and improved ataxia and cerebellar Purkinje cell firing rate deficits. A TrkB agonist, 7,8-dihydroxyflavone (7,8-DHF), likewise improved motor coordination and Purkinje cell firing rate and elevated downstream Akt signaling. Prolonged 7,8-DHF administration persistently improved ataxia when treatment commenced near disease onset but was ineffective when treatment was started late. These data suggest that 7,8-DHF, which is orally bioavailable and crosses the blood-brain barrier, is a promising therapeutic for SCA6 and argue for the importance of early intervention for SCA6.
Collapse
Affiliation(s)
- Anna A. Cook
- Biology Department, McGill University, Montreal, QC, Canada
| | - Sriram Jayabal
- Biology Department, McGill University, Montreal, QC, Canada
- Integrated Neuroscience Program, McGill University, Montreal, QC, Canada
- Department of Neurobiology, Stanford School of Medicine, Stanford, CA, USA
| | - Jacky Sheng
- Biology Department, McGill University, Montreal, QC, Canada
| | - Eviatar Fields
- Biology Department, McGill University, Montreal, QC, Canada
- Integrated Neuroscience Program, McGill University, Montreal, QC, Canada
| | | | - Sabrina Quilez
- Biology Department, McGill University, Montreal, QC, Canada
| | | | - Lois Lau
- Biology Department, McGill University, Montreal, QC, Canada
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Department of Education, Innovation and Technology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Alanna J. Watt
- Biology Department, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
Balbo I, Montarolo F, Genovese F, Tempia F, Hoxha E. Effects of the administration of Elovl5-dependent fatty acids on a spino-cerebellar ataxia 38 mouse model. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2022; 18:8. [PMID: 35933444 PMCID: PMC9357323 DOI: 10.1186/s12993-022-00194-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022]
Abstract
Background Spinocerebellar ataxia 38 (SCA38) is a rare autosomal neurological disorder characterized by ataxia and cerebellar atrophy. SCA38 is caused by mutations of ELOVL5 gene. ELOVL5 gene encodes a protein, which elongates long chain polyunsaturated fatty acids (PUFAs). Knockout mice lacking Elovl5 recapitulate SCA38 symptoms, including motor coordination impairment and disruption of cerebellar architecture. We asked whether, in Elovl5 knockout mice (Elovl5−/−), a diet with both ω3 and ω6 PUFAs downstream Elovl5 can prevent the development of SCA38 symptoms, and at which age such treatment is more effective. Elovl5−/− mice were fed either with a diet without or containing PUFAs downstream the Elovl5 enzyme, starting at different ages. Motor behavior was assessed by the balance beam test and cerebellar structure by morphometric analysis. Results The administration from birth of the diet containing PUFAs downstream Elovl5 led to a significant amelioration of the motor performance in the beam test of Elovl5−/− mice, with a reduction of foot slip errors at 6 months from 2.2 ± 0.3 to 1.3 ± 0.2 and at 8 months from 3.1 ± 0.5 to 1.9 ± 0.3. On the contrary, administration at 1 month of age or later had no effect on the motor impairment. The cerebellar Purkinje cell layer and the white matter area of Elovl5−/ −mice were not rescued even by the administration of diet from birth, suggesting that the improvement of motor performance in the beam test was due to a functional recovery of the cerebellar circuitry. Conclusions These results suggest that the dietary intervention in SCA38, whenever possible, should be started from birth or as early as possible.
Collapse
Affiliation(s)
- Ilaria Balbo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043, Orbassano, Italy
| | - Francesca Montarolo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043, Orbassano, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Federica Genovese
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043, Orbassano, Italy
| | - Filippo Tempia
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043, Orbassano, Italy.,Department of Neuroscience, University of Torino, Torino, Italy
| | - Eriola Hoxha
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole 10, 10043, Orbassano, Italy. .,Department of Neuroscience, University of Torino, Torino, Italy.
| |
Collapse
|
28
|
Li M, Liu F, Hao X, Fan Y, Li J, Hu Z, Shi J, Fan L, Zhang S, Ma D, Guo M, Xu Y, Shi C. Rare KCND3 Loss-of-Function Mutation Associated With the SCA19/22. Front Mol Neurosci 2022; 15:919199. [PMID: 35813061 PMCID: PMC9261871 DOI: 10.3389/fnmol.2022.919199] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/19/2022] [Indexed: 12/15/2022] Open
Abstract
Spinocerebellar ataxia 19/22 (SCA19/22) is a rare neurodegenerative disorder caused by mutations of the KCND3 gene, which encodes the Kv4. 3 protein. Currently, only 22 KCND3 single-nucleotide mutation sites of SCA19/22 have been reported worldwide, and detailed pathogenesis remains unclear. In this study, Sanger sequencing was used to screen 115 probands of cerebellar ataxia families in 67 patients with sporadic cerebellar ataxia and 200 healthy people to identify KCND3 mutations. Mutant gene products showed pathogenicity damage, and the polarity was changed. Next, we established induced pluripotent stem cells (iPSCs) derived from SCA19/22 patients. Using a transcriptome sequencing technique, we found that protein processing in the endoplasmic reticulum was significantly enriched in SCA19/22-iPS-derived neurons and was closely related to endoplasmic reticulum stress (ERS) and apoptosis. In addition, Western blotting of the SCA19/22-iPS-derived neurons showed a reduction in Kv4.3; but, activation of transcription factor 4 (ATF4) and C/EBP homologous protein was increased. Therefore, the c.1130 C>T (p.T377M) mutation of the KCND3 gene may mediate misfold and aggregation of Kv4.3, which activates the ERS and further induces neuron apoptosis involved in SCA19/22.
Collapse
Affiliation(s)
- Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Fen Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Yu Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Jiadi Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Jingjing Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Department of Cell Biology and Medical Genetics, Basic Medical College of Zhengzhou University, Zhengzhou, China
| | - Liyuan Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Dongrui Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Mengnan Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Department of Cell Biology and Medical Genetics, Basic Medical College of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Hereditary Neurodegenerative Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Key Laboratory of Cerebrovascular Diseases Prevention and Treatment, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- The Henan Medical Key Laboratory of Hereditary Neurodegenerative Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Key Laboratory of Cerebrovascular Diseases Prevention and Treatment, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- *Correspondence: Changhe Shi
| |
Collapse
|
29
|
Parvez MSA, Ohtsuki G. Acute Cerebellar Inflammation and Related Ataxia: Mechanisms and Pathophysiology. Brain Sci 2022; 12:367. [PMID: 35326323 PMCID: PMC8946185 DOI: 10.3390/brainsci12030367] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/11/2022] Open
Abstract
The cerebellum governs motor coordination and motor learning. Infection with external microorganisms, such as viruses, bacteria, and fungi, induces the release and production of inflammatory mediators, which drive acute cerebellar inflammation. The clinical observation of acute cerebellitis is associated with the emergence of cerebellar ataxia. In our animal model of the acute inflammation of the cerebellar cortex, animals did not show any ataxia but hyperexcitability in the cerebellar cortex and depression-like behaviors. In contrast, animal models with neurodegeneration of the cerebellar Purkinje cells and hypoexcitability of the neurons show cerebellar ataxia. The suppression of the Ca2+-activated K+ channels in vivo is associated with a type of ataxia. Therefore, there is a gap in our interpretation between the very early phase of cerebellar inflammation and the emergence of cerebellar ataxia. In this review, we discuss the hypothesized scenario concerning the emergence of cerebellar ataxia. First, compared with genetically induced cerebellar ataxias, we introduce infection and inflammation in the cerebellum via aberrant immunity and glial responses. Especially, we focus on infections with cytomegalovirus, influenza virus, dengue virus, and SARS-CoV-2, potential relevance to mitochondrial DNA, and autoimmunity in infection. Second, we review neurophysiological modulation (intrinsic excitability, excitatory, and inhibitory synaptic transmission) by inflammatory mediators and aberrant immunity. Next, we discuss the cerebellar circuit dysfunction (presumably, via maintaining the homeostatic property). Lastly, we propose the mechanism of the cerebellar ataxia and possible treatments for the ataxia in the cerebellar inflammation.
Collapse
Affiliation(s)
- Md. Sorwer Alam Parvez
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8397, Japan;
- Department of Genetic Engineering & Biotechnology, Shahjalal University of Science & Technology, Sylhet 3114, Bangladesh
| | - Gen Ohtsuki
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 606-8397, Japan;
| |
Collapse
|
30
|
Perez H, Abdallah MF, Chavira JI, Norris AS, Egeland MT, Vo KL, Buechsenschuetz CL, Sanghez V, Kim JL, Pind M, Nakamura K, Hicks GG, Gatti RA, Madrenas J, Iacovino M, McKinnon PJ, Mathews PJ. A novel, ataxic mouse model of ataxia telangiectasia caused by a clinically relevant nonsense mutation. eLife 2021; 10:e64695. [PMID: 34723800 PMCID: PMC8601662 DOI: 10.7554/elife.64695] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 10/29/2021] [Indexed: 12/14/2022] Open
Abstract
Ataxia Telangiectasia (A-T) and Ataxia with Ocular Apraxia Type 1 (AOA1) are devastating neurological disorders caused by null mutations in the genome stability genes, A-T mutated (ATM) and Aprataxin (APTX), respectively. Our mechanistic understanding and therapeutic repertoire for treating these disorders are severely lacking, in large part due to the failure of prior animal models with similar null mutations to recapitulate the characteristic loss of motor coordination (i.e., ataxia) and associated cerebellar defects. By increasing genotoxic stress through the insertion of null mutations in both the Atm (nonsense) and Aptx (knockout) genes in the same animal, we have generated a novel mouse model that for the first time develops a progressively severe ataxic phenotype associated with atrophy of the cerebellar molecular layer. We find biophysical properties of cerebellar Purkinje neurons (PNs) are significantly perturbed (e.g., reduced membrane capacitance, lower action potential [AP] thresholds, etc.), while properties of synaptic inputs remain largely unchanged. These perturbations significantly alter PN neural activity, including a progressive reduction in spontaneous AP firing frequency that correlates with both cerebellar atrophy and ataxia over the animal's first year of life. Double mutant mice also exhibit a high predisposition to developing cancer (thymomas) and immune abnormalities (impaired early thymocyte development and T-cell maturation), symptoms characteristic of A-T. Finally, by inserting a clinically relevant nonsense-type null mutation in Atm, we demonstrate that Small Molecule Read-Through (SMRT) compounds can restore ATM production, indicating their potential as a future A-T therapeutic.
Collapse
Affiliation(s)
- Harvey Perez
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
| | - May F Abdallah
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
| | - Jose I Chavira
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
| | - Angelina S Norris
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
| | - Martin T Egeland
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
| | - Karen L Vo
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
| | - Callan L Buechsenschuetz
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
| | - Valentina Sanghez
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
| | - Jeannie L Kim
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
| | - Molly Pind
- Department of Biochemistry and Medical Genetics,Max Rady College of Medicine, University of ManitobaManitobaCanada
| | - Kotoka Nakamura
- Department of Pathology & Laboratory Medicine, David Geffen School of MedicineLos AngelesUnited States
| | - Geoffrey G Hicks
- Department of Biochemistry and Medical Genetics,Max Rady College of Medicine, University of ManitobaManitobaCanada
| | - Richard A Gatti
- Department of Pathology & Laboratory Medicine, David Geffen School of MedicineLos AngelesUnited States
| | - Joaquin Madrenas
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
- Department of Medicine, Harbor-UCLA Medical CenterTorranceUnited States
| | - Michelina Iacovino
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
- Department of Pediatrics, Harbor-UCLA Medical CenterTorranceUnited States
| | - Peter J McKinnon
- Center for Pediatric Neurological Disease Research, St. Jude Pediatric Translational Neuroscience Initiative, St. Jude Children’s Research HospitalMemphisUnited States
| | - Paul J Mathews
- The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical CenterTorranceUnited States
- Department of Neurology, Harbor-UCLA Medical CenterTorranceUnited States
| |
Collapse
|
31
|
Borbolla-Jiménez FV, Del Prado-Audelo ML, Cisneros B, Caballero-Florán IH, Leyva-Gómez G, Magaña JJ. New Perspectives of Gene Therapy on Polyglutamine Spinocerebellar Ataxias: From Molecular Targets to Novel Nanovectors. Pharmaceutics 2021; 13:1018. [PMID: 34371710 PMCID: PMC8309146 DOI: 10.3390/pharmaceutics13071018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 01/03/2023] Open
Abstract
Seven of the most frequent spinocerebellar ataxias (SCAs) are caused by a pathological expansion of a cytosine, adenine and guanine (CAG) trinucleotide repeat located in exonic regions of unrelated genes, which in turn leads to the synthesis of polyglutamine (polyQ) proteins. PolyQ proteins are prone to aggregate and form intracellular inclusions, which alter diverse cellular pathways, including transcriptional regulation, protein clearance, calcium homeostasis and apoptosis, ultimately leading to neurodegeneration. At present, treatment for SCAs is limited to symptomatic intervention, and there is no therapeutic approach to prevent or reverse disease progression. This review provides a compilation of the experimental advances obtained in cell-based and animal models toward the development of gene therapy strategies against polyQ SCAs, providing a discussion of their potential application in clinical trials. In the second part, we describe the promising potential of nanotechnology developments to treat polyQ SCA diseases. We describe, in detail, how the design of nanoparticle (NP) systems with different physicochemical and functionalization characteristics has been approached, in order to determine their ability to evade the immune system response and to enhance brain delivery of molecular tools. In the final part of this review, the imminent application of NP-based strategies in clinical trials for the treatment of polyQ SCA diseases is discussed.
Collapse
Affiliation(s)
- Fabiola V. Borbolla-Jiménez
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de México 14389, Mexico;
- Programa de Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - María Luisa Del Prado-Audelo
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey Campus Ciudad de México, Ciudad de México 14380, Mexico;
| | - Bulmaro Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México 07360, Mexico;
| | - Isaac H. Caballero-Florán
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
- Departamento de Farmacia, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México 07360, Mexico
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Jonathan J. Magaña
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de México 14389, Mexico;
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey Campus Ciudad de México, Ciudad de México 14380, Mexico;
| |
Collapse
|
32
|
Hommersom MP, Buijsen RAM, van Roon-Mom WMC, van de Warrenburg BPC, van Bokhoven H. Human Induced Pluripotent Stem Cell-Based Modelling of Spinocerebellar Ataxias. Stem Cell Rev Rep 2021; 18:441-456. [PMID: 34031815 PMCID: PMC8930896 DOI: 10.1007/s12015-021-10184-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Abstract Dominant spinocerebellar ataxias (SCAs) constitute a large group of phenotypically and genetically heterogeneous disorders that mainly present with dysfunction of the cerebellum as their main hallmark. Although animal and cell models have been highly instrumental for our current insight into the underlying disease mechanisms of these neurodegenerative disorders, they do not offer the full human genetic and physiological context. The advent of human induced pluripotent stem cells (hiPSCs) and protocols to differentiate these into essentially every cell type allows us to closely model SCAs in a human context. In this review, we systematically summarize recent findings from studies using hiPSC-based modelling of SCAs, and discuss what knowledge has been gained from these studies. We conclude that hiPSC-based models are a powerful tool for modelling SCAs as they contributed to new mechanistic insights and have the potential to serve the development of genetic therapies. However, the use of standardized methods and multiple clones of isogenic lines are essential to increase validity and reproducibility of the insights gained. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Marina P Hommersom
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Ronald A M Buijsen
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Willeke M C van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Bart P C van de Warrenburg
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.
| | - Hans van Bokhoven
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands. .,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6500 HB, Nijmegen, Netherlands.
| |
Collapse
|
33
|
van der Heijden ME, Kizek DJ, Perez R, Ruff EK, Ehrlich ME, Sillitoe RV. Abnormal cerebellar function and tremor in a mouse model for non-manifesting partially penetrant dystonia type 6. J Physiol 2021; 599:2037-2054. [PMID: 33369735 PMCID: PMC8559601 DOI: 10.1113/jp280978] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/16/2020] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Loss-of-function mutations in the Thap1 gene cause partially penetrant dystonia type 6 (DYT6). Some non-manifesting DYT6 mutation carriers have tremor and abnormal cerebello-thalamo-cortical signalling. We show that Thap1 heterozygote mice have action tremor, a reduction in cerebellar neuron number, and abnormal electrophysiological signals in the remaining neurons. These results underscore the importance of Thap1 levels for cerebellar function. These results uncover how cerebellar abnormalities contribute to different dystonia-associated motor symptoms. ABSTRACT Loss-of-function mutations in the Thanatos-associated domain-containing apoptosis-associated protein 1 (THAP1) gene cause partially penetrant autosomal dominant dystonia type 6 (DYT6). However, the neural abnormalities that promote the resultant motor dysfunctions remain elusive. Studies in humans show that some non-manifesting DYT6 carriers have altered cerebello-thalamo-cortical function with subtle but reproducible tremor. Here, we uncover that Thap1 heterozygote mice have action tremor that rises above normal baseline values even though they do not exhibit overt dystonia-like twisting behaviour. At the neural circuit level, we show using in vivo recordings in awake Thap1+/- mice that Purkinje cells have abnormal firing patterns and that cerebellar nuclei neurons, which connect the cerebellum to the thalamus, fire at a lower frequency. Although the Thap1+/- mice have fewer Purkinje cells and cerebellar nuclei neurons, the number of long-range excitatory outflow projection neurons is unaltered. The preservation of interregional connectivity suggests that abnormal neural function rather than neuron loss instigates the network dysfunction and the tremor in Thap1+/- mice. Accordingly, we report an inverse correlation between the average firing rate of cerebellar nuclei neurons and tremor power. Our data show that cerebellar circuitry is vulnerable to Thap1 mutations and that cerebellar dysfunction may be a primary cause of tremor in non-manifesting DYT6 carriers and a trigger for the abnormal postures in manifesting patients.
Collapse
Affiliation(s)
- Meike E. van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Dominic J. Kizek
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Ross Perez
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Elena K. Ruff
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Michelle E. Ehrlich
- Department of Neurology and Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, 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
- Program in Developmental Biology, 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, USA
| |
Collapse
|
34
|
Hooshmandi M, Truong VT, Fields E, Thomas RE, Wong C, Sharma V, Gantois I, Soriano Roque P, Chalkiadaki K, Wu N, Chakraborty A, Tahmasebi S, Prager-Khoutorsky M, Sonenberg N, Suvrathan A, Watt AJ, Gkogkas CG, Khoutorsky A. 4E-BP2-dependent translation in cerebellar Purkinje cells controls spatial memory but not autism-like behaviors. Cell Rep 2021; 35:109036. [PMID: 33910008 DOI: 10.1016/j.celrep.2021.109036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/15/2021] [Accepted: 04/06/2021] [Indexed: 11/19/2022] Open
Abstract
Recent studies have demonstrated that selective activation of mammalian target of rapamycin complex 1 (mTORC1) in the cerebellum by deletion of the mTORC1 upstream repressors TSC1 or phosphatase and tensin homolog (PTEN) in Purkinje cells (PCs) causes autism-like features and cognitive deficits. However, the molecular mechanisms by which overactivated mTORC1 in the cerebellum engenders these behaviors remain unknown. The eukaryotic translation initiation factor 4E-binding protein 2 (4E-BP2) is a central translational repressor downstream of mTORC1. Here, we show that mice with selective ablation of 4E-BP2 in PCs display a reduced number of PCs, increased regularity of PC action potential firing, and deficits in motor learning. Surprisingly, although spatial memory is impaired in these mice, they exhibit normal social interaction and show no deficits in repetitive behavior. Our data suggest that, downstream of mTORC1/4E-BP2, there are distinct cerebellar mechanisms independently controlling social behavior and memory formation.
Collapse
Affiliation(s)
- Mehdi Hooshmandi
- Department of Anesthesia and Faculty of Dentistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Vinh Tai Truong
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Eviatar Fields
- Department of Biology, McGill University, Montreal, QC H3A 1A3, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B4, Canada
| | - Riya Elizabeth Thomas
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B4, Canada; Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal QC, H3G1A4, Canada; Department of Neurology and Neurosurgery, Department of Pediatrics, McGill University, Montreal QC, H3G1A4, Canada
| | - Calvin Wong
- Department of Anesthesia and Faculty of Dentistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Vijendra Sharma
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Ilse Gantois
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Patricia Soriano Roque
- Department of Anesthesia and Faculty of Dentistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Kleanthi Chalkiadaki
- Division of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
| | - Neil Wu
- Department of Anesthesia and Faculty of Dentistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Anindyo Chakraborty
- Department of Anesthesia and Faculty of Dentistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Soroush Tahmasebi
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Aparna Suvrathan
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montreal QC, H3G1A4, Canada; Department of Neurology and Neurosurgery, Department of Pediatrics, McGill University, Montreal QC, H3G1A4, Canada
| | - Alanna J Watt
- Department of Biology, McGill University, Montreal, QC H3A 1A3, Canada
| | - Christos G Gkogkas
- Division of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece.
| | - Arkady Khoutorsky
- Department of Anesthesia and Faculty of Dentistry, McGill University, Montreal, QC H3G 1Y6, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC H3A 0G1, Canada.
| |
Collapse
|
35
|
Sheeler C, Rosa JG, Borgenheimer E, Mellesmoen A, Rainwater O, Cvetanovic M. Post-symptomatic Delivery of Brain-Derived Neurotrophic Factor (BDNF) Ameliorates Spinocerebellar Ataxia Type 1 (SCA1) Pathogenesis. THE CEREBELLUM 2021; 20:420-429. [PMID: 33394333 DOI: 10.1007/s12311-020-01226-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/06/2020] [Indexed: 11/26/2022]
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an abnormal expansion of CAG repeats in the Ataxin1 (ATXN1) gene. SCA1 is characterized by motor deficits, cerebellar neurodegeneration, and gliosis and gene expression changes. Expression of brain-derived neurotrophic factor (BDNF), growth factor important for the survival and function of cerebellar neurons, is decreased in ATXN1[82Q] mice, the Purkinje neuron specific transgenic mouse model of SCA1. As this decrease in BDNF expression may contribute to cerebellar neurodegeneration, we tested whether delivery of extrinsic human BDNF via osmotic ALZET pumps has a beneficial effect on disease severity in this mouse model of SCA1. Additionally, to test the effects of BDNF on established and progressing cerebellar pathogenesis and motor deficits, we delivered BDNF post-symptomatically. We have found that post-symptomatic delivery of extrinsic BDNF ameliorated motor deficits and cerebellar pathology (i.e., dendritic atrophy of Purkinje cells, and astrogliosis) indicating therapeutic potential of BDNF even after the onset of symptoms in SCA1. However, BDNF did not alter Purkinje cell gene expression changes indicating that certain aspects of disease pathogenesis cannot be ameliorated/slowed down with BDNF and that combinational therapies may be needed.
Collapse
Affiliation(s)
- Carrie Sheeler
- Department of Neuroscience, University of Minnesota, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Juao-Guilherme Rosa
- Department of Neuroscience, University of Minnesota, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Ella Borgenheimer
- Department of Neuroscience, University of Minnesota, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Aaron Mellesmoen
- Department of Neuroscience, University of Minnesota, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Orion Rainwater
- Department of Lab Medicine and Pathology, University of Minnesota, 420 Delaware Street SE, Minneapolis, 55455, USA
| | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, 321 Church Street SE, Minneapolis, MN, 55455, USA.
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
36
|
Chopra R, Bushart DD, Cooper JP, Yellajoshyula D, Morrison LM, Huang H, Handler HP, Man LJ, Dansithong W, Scoles DR, Pulst SM, Orr HT, Shakkottai VG. Altered Capicua expression drives regional Purkinje neuron vulnerability through ion channel gene dysregulation in spinocerebellar ataxia type 1. Hum Mol Genet 2020; 29:3249-3265. [PMID: 32964235 PMCID: PMC7689299 DOI: 10.1093/hmg/ddaa212] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/19/2020] [Accepted: 09/17/2020] [Indexed: 12/22/2022] Open
Abstract
Selective neuronal vulnerability in neurodegenerative disease is poorly understood. Using the ATXN1[82Q] model of spinocerebellar ataxia type 1 (SCA1), we explored the hypothesis that regional differences in Purkinje neuron degeneration could provide novel insights into selective vulnerability. ATXN1[82Q] Purkinje neurons from the anterior cerebellum were found to degenerate earlier than those from the nodular zone, and this early degeneration was associated with selective dysregulation of ion channel transcripts and altered Purkinje neuron spiking. Efforts to understand the basis for selective dysregulation of channel transcripts revealed modestly increased expression of the ATXN1 co-repressor Capicua (Cic) in anterior cerebellar Purkinje neurons. Importantly, disrupting the association between ATXN1 and Cic rescued the levels of these ion channel transcripts, and lentiviral overexpression of Cic in the nodular zone accelerated both aberrant Purkinje neuron spiking and neurodegeneration. These findings reinforce the central role for Cic in SCA1 cerebellar pathophysiology and suggest that only modest reductions in Cic are needed to have profound therapeutic impact in SCA1.
Collapse
Affiliation(s)
- Ravi Chopra
- Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Neurology, Washington University in St. Louis, Saint Louis, MO 63110, USA
| | - David D Bushart
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - John P Cooper
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | | | - Logan M Morrison
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Haoran Huang
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hillary P Handler
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Luke J Man
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Warunee Dansithong
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Daniel R Scoles
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Harry T Orr
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vikram G Shakkottai
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
37
|
Bushart DD, Huang H, Man LJ, Morrison LM, Shakkottai VG. A Chlorzoxazone-Baclofen Combination Improves Cerebellar Impairment in Spinocerebellar Ataxia Type 1. Mov Disord 2020; 36:622-631. [PMID: 33151010 DOI: 10.1002/mds.28355] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A combination of central muscle relaxants, chlorzoxazone and baclofen (chlorzoxazone-baclofen), has been proposed for treatment of cerebellar symptoms in human spinocerebellar ataxia. However, central muscle relaxants can worsen balance. The optimal dose for target engagement without toxicity remains unknown. Using the genetically precise Atxn1154Q/2Q model of spinocerebellar ataxia type 1, we aimed to determine the role of cerebellar dysfunction in motor impairment. We also aimed to identify appropriate concentrations of chlorzoxazone-baclofen needed for target engagement without toxicity to plan for human clinical trials. METHODS We use patch clamp electrophysiology in acute cerebellar slices and immunostaining to identify the specific ion channels targeted by chlorzoxazone-baclofen. Behavioral assays for coordination and grip strength are used to determine specificity of chlorzoxazone-baclofen for improving cerebellar dysfunction without off-target effects in Atxn1154Q/2Q mice. RESULTS We identify irregular Purkinje neuron firing in association with reduced expression of ion channels Kcnma1 and Cacna1g in Atxn1154Q/2Q mice. Using in vitro electrophysiology in brain slices, we identified concentrations of chlorzoxazone-baclofen that improve Purkinje neuron spike regularity without reducing firing frequency. At a disease stage in Atxn1154Q/2Q mice when motor impairment is due to cerebellar dysfunction, orally administered chlorzoxazone-baclofen improves motor performance without affecting muscle strength. CONCLUSION We identify a tight relationship between baclofen-chlorzoxazone concentrations needed to engage target and levels above which cerebellar function will be compromised. We propose to use this information for a novel clinical trial design, using sequential dose escalation within each subject, to identify dose levels that are likely to improve ataxia symptoms while minimizing toxicity. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- David D Bushart
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Haoran Huang
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Luke J Man
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Logan M Morrison
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Vikram G Shakkottai
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
38
|
Kiven S, Wang Y, Aich A, Argueta DA, Lei J, Sagi V, Tennakoon M, Bedros SJ, Lambrecht N, Gupta K. Spatiotemporal Alterations in Gait in Humanized Transgenic Sickle Mice. Front Immunol 2020; 11:561947. [PMID: 33178189 PMCID: PMC7593487 DOI: 10.3389/fimmu.2020.561947] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
Sickle cell disease (SCD) is a hemoglobinopathy affecting multiple organs and featuring acute and chronic pain. Purkinje cell damage and hyperalgesia have been demonstrated in transgenic sickle mice. Purkinje cells are associated with movement and neural function which may influence pain. We hypothesized that Purkinje cell damage and/or chronic pain burden provoke compensatory gait changes in sickle mice. We found that Purkinje cells undergoe increased apoptosis as shown by caspase-3 activation. Using an automated gait measurement system, MouseWalker, we characterized spatiotemporal gait characteristics of humanized transgenic BERK sickle mice in comparison to control mice. Sickle mice showed alteration in stance instability and dynamic gait parameters (walking speed, stance duration, swing duration and specific swing indices). Differences in stance instability may reflect motor dysfunction due to damaged Purkinje cells. Alterations in diagonal and all stance indices indicative of hesitation during walking may originate from motor dysfunction and/or arise from fear and/or anticipation of movement-evoked pain. We also demonstrate that stance duration, diagonal swing indices and all stance indices correlate with both mechanical and deep tissue hyperalgesia, while stance instability correlates with only deep tissue hyperalgesia. Therefore, objective analysis of gait in SCD may provide insights into neurological impairment and pain states.
Collapse
Affiliation(s)
- Stacy Kiven
- Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
| | - Ying Wang
- Department of Anesthesia, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anupam Aich
- Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Donovan A. Argueta
- Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Jianxun Lei
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
| | - Varun Sagi
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
| | - Madhushan Tennakoon
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
| | - Saad J. Bedros
- College of Science & Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Nils Lambrecht
- Pathology and Laboratory Medicine, Long Beach VA Healthcare System, Long Beach, CA, United States
| | - Kalpna Gupta
- Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN, United States
- Southern California Institute for Research and Education, Long Beach VA Healthcare System, Long Beach, CA, United States
| |
Collapse
|
39
|
Neves-Carvalho A, Duarte-Silva S, Teixeira-Castro A, Maciel P. Polyglutamine spinocerebellar ataxias: emerging therapeutic targets. Expert Opin Ther Targets 2020; 24:1099-1119. [PMID: 32962458 DOI: 10.1080/14728222.2020.1827394] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Six of the most frequent dominantly inherited spinocerebellar ataxias (SCAs) worldwide - SCA1, SCA2, SCA3, SCA6, SCA7, and SCA17 - are caused by an expansion of a polyglutamine (polyQ) tract in the corresponding proteins. While the identification of the causative mutation has advanced knowledge on the pathogenesis of polyQ SCAs, effective therapeutics able to mitigate the severe clinical manifestation of these highly incapacitating disorders are not yet available. AREAS COVERED This review provides a comprehensive and critical perspective on well-established and emerging therapeutic targets for polyQ SCAs; it aims to inspire prospective drug discovery efforts. EXPERT OPINION The landscape of polyQ SCAs therapeutic targets and strategies includes (1) the mutant genes and proteins themselves, (2) enhancement of endogenous protein quality control responses, (3) abnormal protein-protein interactions of the mutant proteins, (4) disturbed neuronal function, (5) mitochondrial function, energy availability and oxidative stress, and (6) glial dysfunction, growth factor or hormone imbalances. Challenges include gaining a clearer definition of therapeutic targets for the drugs in clinical development, the discovery of novel drug-like molecules for challenging key targets, and the attainment of a stronger translation of preclinical findings to the clinic.
Collapse
Affiliation(s)
- Andreia Neves-Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| |
Collapse
|
40
|
Robinson KJ, Watchon M, Laird AS. Aberrant Cerebellar Circuitry in the Spinocerebellar Ataxias. Front Neurosci 2020; 14:707. [PMID: 32765211 PMCID: PMC7378801 DOI: 10.3389/fnins.2020.00707] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022] Open
Abstract
The spinocerebellar ataxias (SCAs) are a heterogeneous group of neurodegenerative diseases that share convergent disease features. A common symptom of these diseases is development of ataxia, involving impaired balance and motor coordination, usually stemming from cerebellar dysfunction and neurodegeneration. For most spinocerebellar ataxias, pathology can be attributed to an underlying gene mutation and the impaired function of the encoded protein through loss or gain-of-function effects. Strikingly, despite vast heterogeneity in the structure and function of disease-causing genes across the SCAs and the cellular processes affected, the downstream effects have considerable overlap, including alterations in cerebellar circuitry. Interestingly, aberrant function and degeneration of Purkinje cells, the major output neuronal population present within the cerebellum, precedes abnormalities in other neuronal populations within many SCAs, suggesting that Purkinje cells have increased vulnerability to cellular perturbations. Factors that are known to contribute to perturbed Purkinje cell function in spinocerebellar ataxias include altered gene expression resulting in altered expression or functionality of proteins and channels that modulate membrane potential, downstream impairments in intracellular calcium homeostasis and changes in glutamatergic input received from synapsing climbing or parallel fibers. This review will explore this enhanced vulnerability and the aberrant cerebellar circuitry linked with it in many forms of SCA. It is critical to understand why Purkinje cells are vulnerable to such insults and what overlapping pathogenic mechanisms are occurring across multiple SCAs, despite different underlying genetic mutations. Enhanced understanding of disease mechanisms will facilitate the development of treatments to prevent or slow progression of the underlying neurodegenerative processes, cerebellar atrophy and ataxic symptoms.
Collapse
Affiliation(s)
| | | | - Angela S. Laird
- Centre for Motor Neuron Disease Research, Department of Biomedical Science, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
41
|
Cook AA, Fields E, Watt AJ. Losing the Beat: Contribution of Purkinje Cell Firing Dysfunction to Disease, and Its Reversal. Neuroscience 2020; 462:247-261. [PMID: 32554108 DOI: 10.1016/j.neuroscience.2020.06.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023]
Abstract
The cerebellum is a brain structure that is highly interconnected with other brain regions. There are many contributing factors to cerebellar-related brain disease, such as altered afferent input, local connectivity, and/or cerebellar output. Purkinje cells (PC) are the principle cells of the cerebellar cortex, and fire intrinsically; that is, they fire spontaneous action potentials at high frequencies. This review paper focuses on PC intrinsic firing activity, which is altered in multiple neurological diseases, including ataxia, Huntington Disease (HD) and autism spectrum disorder (ASD). Notably, there are several cases where interventions that restore or rescue PC intrinsic activity also improve impaired behavior in these mouse models of disease. These findings suggest that rescuing PC firing deficits themselves may be sufficient to improve impairment in cerebellar-related behavior in disease. We propose that restoring PC intrinsic firing represents a good target for drug development that might be of therapeutic use for several disorders.
Collapse
Affiliation(s)
- Anna A Cook
- Department of Biology, McGill University, Montreal, Canada
| | - Eviatar Fields
- Department of Biology, McGill University, Montreal, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Alanna J Watt
- Department of Biology, McGill University, Montreal, Canada.
| |
Collapse
|
42
|
Binda F, Pernaci C, Saxena S. Cerebellar Development and Circuit Maturation: A Common Framework for Spinocerebellar Ataxias. Front Neurosci 2020; 14:293. [PMID: 32300292 PMCID: PMC7145357 DOI: 10.3389/fnins.2020.00293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 03/13/2020] [Indexed: 01/24/2023] Open
Abstract
Spinocerebellar ataxias (SCAs) affect the cerebellum and its afferent and efferent systems that degenerate during disease progression. In the cerebellum, Purkinje cells (PCs) are the most vulnerable and their prominent loss in the late phase of the pathology is the main characteristic of these neurodegenerative diseases. Despite the constant advancement in the discovery of affected molecules and cellular pathways, a comprehensive description of the events leading to the development of motor impairment and degeneration is still lacking. However, in the last years the possible causal role for altered cerebellar development and neuronal circuit wiring in SCAs has been emerging. Not only wiring and synaptic transmission deficits are a common trait of SCAs, but also preventing the expression of the mutant protein during cerebellar development seems to exert a protective role. By discussing this tight relationship between cerebellar development and SCAs, in this review, we aim to highlight the importance of cerebellar circuitry for the investigation of SCAs.
Collapse
Affiliation(s)
- Francesca Binda
- Department of Neurology, Center for Experimental Neurology, University Hospital of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Carla Pernaci
- Department of Neurology, Center for Experimental Neurology, University Hospital of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Smita Saxena
- Department of Neurology, Center for Experimental Neurology, University Hospital of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
43
|
Volovikov EA, Davidenko AV, Lagarkova MA. Molecular Mechanisms of Spinocerebellar Ataxia Type 1. RUSS J GENET+ 2020. [DOI: 10.1134/s102279542002012x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
44
|
The neurological update: therapies for cerebellar ataxias in 2020. J Neurol 2020; 267:1211-1220. [DOI: 10.1007/s00415-020-09717-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/12/2020] [Accepted: 01/18/2020] [Indexed: 12/28/2022]
|
45
|
Brown AS, Meera P, Quinones G, Magri J, Otis TS, Pulst SM, Oro AE. Receptor protein tyrosine phosphatases control Purkinje neuron firing. Cell Cycle 2020; 19:153-159. [PMID: 31876231 PMCID: PMC6961678 DOI: 10.1080/15384101.2019.1695995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/23/2019] [Accepted: 11/07/2019] [Indexed: 10/25/2022] Open
Abstract
Spinocerebellar ataxias (SCA) are a genetically heterogeneous family of cerebellar neurodegenerative diseases characterized by abnormal firing of Purkinje neurons and degeneration. We recently demonstrated the slowed firing rates seen in several SCAs share a common etiology of hyper-activation of the Src family of non-receptor tyrosine kinases (SFKs). However, the lack of clinically available neuroactive SFK inhibitors lead us to investigate alternative mechanisms to modulate SFK activity. Previous studies demonstrate that SFK activity can be enhanced by the removal of inhibitory phospho-marks by receptor-protein-tyrosine phosphatases (RPTPs). In this Extra View we show that MTSS1 inhibits SFK activity through the binding and inhibition of a subset of the RPTP family members, and lowering RPTP activity in cerebellar slices with peptide inhibitors increases the suppressed Purkinje neuron basal firing rates seen in two different SCA models. Together these results identify RPTPs as novel effectors of Purkinje neuron basal firing, extending the MTSS1/SFK regulatory circuit we previously described and expanding the therapeutic targets for SCA patients.
Collapse
Affiliation(s)
- Alexander S. Brown
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Pratap Meera
- Department of Neurobiology, University of California, Los Angeles, CA, USA
| | - Gabe Quinones
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jessica Magri
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas S. Otis
- Sainsbury Wellcome Centre for Neural Circuits and Behavior, University College London, London, UK
| | - Stefan M. Pulst
- Department of Neurology, University of Utah Medical Center, Salt Lake City, UT, USA
| | - Anthony E. Oro
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
46
|
Srinivasan SR, Shakkottai VG. Moving Towards Therapy in SCA1: Insights from Molecular Mechanisms, Identification of Novel Targets, and Planning for Human Trials. Neurotherapeutics 2019; 16:999-1008. [PMID: 31338702 PMCID: PMC6985354 DOI: 10.1007/s13311-019-00763-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The spinocerebellar ataxias (SCAs) are a group of neurodegenerative disorders inherited in an autosomal dominant fashion. The SCAs result in progressive gait imbalance, incoordination of the limbs, speech changes, and oculomotor dysfunction, among other symptoms. Over the past few decades, significant strides have been made in understanding the pathogenic mechanisms underlying these diseases. Although multiple efforts using a combination of genetics and pharmacology with small molecules have been made towards developing new therapeutics, no FDA approved treatment currently exists. In this review, we focus on SCA1, a common SCA subtype, in which some of the greatest advances have been made in understanding disease biology, and consequently potential therapeutic targets. Understanding of the underlying basic biology and targets of therapy in SCA1 is likely to give insight into treatment strategies in other SCAs. The diversity of the biology in the SCAs, and insight from SCA1 suggests, however, that both shared treatment strategies and specific approaches tailored to treat distinct genetic causes of SCA are likely needed for this group of devastating neurological disorders.
Collapse
Affiliation(s)
| | - Vikram G Shakkottai
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, 4009 BSRB, 109 Zina Pitcher Place, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
47
|
Tsai PJ, Yeh CC, Huang WJ, Min MY, Huang TH, Ko TL, Huang PY, Chen TH, Hsu SPC, Soong BW, Fu YS. Xenografting of human umbilical mesenchymal stem cells from Wharton's jelly ameliorates mouse spinocerebellar ataxia type 1. Transl Neurodegener 2019; 8:29. [PMID: 31508229 PMCID: PMC6727337 DOI: 10.1186/s40035-019-0166-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 08/05/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Spinocerebellar ataxia type 1 (SCA1) is an autosomal dominant neurodegenerative disorder caused by the expansion of CAG repeats in ATXN1 gene resulting in an expansion of polyglutamine repeats in the ATXN1 protein. Unfortunately, there has yet been any effective treatment so far for SCA1. This study investigated the feasibility of transplanting human umbilical mesenchymal stem cells (HUMSCs) into transgenic SCA1 mice containing an expanded uninterrupted allele with 82 repeats in the ATXN1-coding region. METHODS 106 human umbilical mesenchymal stem cells were transplanted into the cerebella at 1 month of age. RESULTS HUMSCs displayed significant ameliorating effects in SCA1 mice in terms of motor behaviors in balance beam test and open field test as compared with the untransplanted SCA1 mice. HUMSCs transplantation effectively reduced the cerebellar atrophy, salvaged Purkinje cell death, and alleviated molecular layer shrinkage. Electrophysiological studies showed higher amplitudes of compound motor action potentials as indicated by increasing neuronal-muscular response strength to stimuli after stem cell transplantation. At 5 months after transplantation, HUMSCs scattering in the mice cerebella remained viable and secreted cytokines without differentiating into neuronal or glia cells. CONCLUSIONS Our findings provide hope for a new therapeutic direction for the treatment of SCA1.
Collapse
Affiliation(s)
- Pei-Jiun Tsai
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Trauma Center, Department of Surgery, Veterans General Hospital, Taipei, Taiwan, Republic of China
- Department of Critical Care Medicine, Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Chang-Ching Yeh
- Department of Obstetrics and Gynecology, Veterans General Hospital, Taipei, Taiwan, Republic of China
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Wan-Jhen Huang
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Ming-Yuan Min
- Department of Life Science, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Tzu-Hao Huang
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Tsui-Ling Ko
- School of Medicine, I-Shou University, Kaohsiung, Taiwan, Republic of China
| | - Pei-Yu Huang
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Tien-Hua Chen
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
- Trauma Center, Department of Surgery, Veterans General Hospital, Taipei, Taiwan, Republic of China
- Division of General Surgery, Department of Surgery, Veterans General Hospital, Taipei, Taiwan, Republic of China
| | - Sanford P. C. Hsu
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Bing-Wen Soong
- Department of Neurology, Shuang Ho Hospital, and Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan, Republic of China
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Yu-Show Fu
- Department of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, No. 155, Sec. 2, Li-Nung Street, Taipei, 112 Taiwan, Republic of China
| |
Collapse
|
48
|
Abstract
The spinocerebellar ataxias (SCAs) comprise more than 40 autosomal dominant neurodegenerative disorders that present principally with progressive ataxia. Within the past few years, studies of pathogenic mechanisms in the SCAs have led to the development of promising therapeutic strategies, especially for SCAs caused by polyglutamine-coding CAG repeats. Nucleotide-based gene-silencing approaches that target the first steps in the pathogenic cascade are one promising approach not only for polyglutamine SCAs but also for the many other SCAs caused by toxic mutant proteins or RNA. For these and other emerging therapeutic strategies, well-coordinated preparation is needed for fruitful clinical trials. To accomplish this goal, investigators from the United States and Europe are now collaborating to share data from their respective SCA cohorts. Increased knowledge of the natural history of SCAs, including of the premanifest and early symptomatic stages of disease, will improve the prospects for success in clinical trials of disease-modifying drugs. In addition, investigators are seeking validated clinical outcome measures that demonstrate responsiveness to changes in SCA populations. Findings suggest that MRI and magnetic resonance spectroscopy biomarkers will provide objective biological readouts of disease activity and progression, but more work is needed to establish disease-specific biomarkers that track target engagement in therapeutic trials. Together, these efforts suggest that the development of successful therapies for one or more SCAs is not far away.
Collapse
|
49
|
Geminiani A, Casellato C, D'Angelo E, Pedrocchi A. Complex Electroresponsive Dynamics in Olivocerebellar Neurons Represented With Extended-Generalized Leaky Integrate and Fire Models. Front Comput Neurosci 2019; 13:35. [PMID: 31244635 PMCID: PMC6563830 DOI: 10.3389/fncom.2019.00035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/20/2019] [Indexed: 11/24/2022] Open
Abstract
The neurons of the olivocerebellar circuit exhibit complex electroresponsive dynamics, which are thought to play a fundamental role for network entraining, plasticity induction, signal processing, and noise filtering. In order to reproduce these properties in single-point neuron models, we have optimized the Extended-Generalized Leaky Integrate and Fire (E-GLIF) neuron through a multi-objective gradient-based algorithm targeting the desired input–output relationships. In this way, E-GLIF was tuned toward the unique input–output properties of Golgi cells, granule cells, Purkinje cells, molecular layer interneurons, deep cerebellar nuclei cells, and inferior olivary cells. E-GLIF proved able to simulate the complex cell-specific electroresponsive dynamics of the main olivocerebellar neurons including pacemaking, adaptation, bursting, post-inhibitory rebound excitation, subthreshold oscillations, resonance, and phase reset. The integration of these E-GLIF point-neuron models into olivocerebellar Spiking Neural Networks will allow to evaluate the impact of complex electroresponsive dynamics at the higher scales, up to motor behavior, in closed-loop simulations of sensorimotor tasks.
Collapse
Affiliation(s)
- Alice Geminiani
- NEARLab, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Claudia Casellato
- 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
| | - Alessandra Pedrocchi
- NEARLab, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
50
|
Abstract
The spinocerebellar ataxias (SCAs) are a genetically heterogeneous group of autosomal dominantly inherited progressive disorders, the clinical hallmark of which is loss of balance and coordination accompanied by slurred speech; onset is most often in adult life. Genetically, SCAs are grouped as repeat expansion SCAs, such as SCA3/Machado-Joseph disease (MJD), and rare SCAs that are caused by non-repeat mutations, such as SCA5. Most SCA mutations cause prominent damage to cerebellar Purkinje neurons with consecutive cerebellar atrophy, although Purkinje neurons are only mildly affected in some SCAs. Furthermore, other parts of the nervous system, such as the spinal cord, basal ganglia and pontine nuclei in the brainstem, can be involved. As there is currently no treatment to slow or halt SCAs (many SCAs lead to premature death), the clinical care of patients with SCA focuses on managing the symptoms through physiotherapy, occupational therapy and speech therapy. Intense research has greatly expanded our understanding of the pathobiology of many SCAs, revealing that they occur via interrelated mechanisms (including proteotoxicity, RNA toxicity and ion channel dysfunction), and has led to the identification of new targets for treatment development. However, the development of effective therapies is hampered by the heterogeneity of the SCAs; specific therapeutic approaches may be required for each disease.
Collapse
|