1
|
Sheeler C, Labrada E, Duvick L, Thompson LM, Zhang Y, Orr HT, Cvetanovic M. Expanded ATXN1 alters transcription and calcium signaling in SCA1 human motor neurons differentiated from induced pluripotent stem cells. Neurobiol Dis 2024; 201:106673. [PMID: 39307401 PMCID: PMC11514977 DOI: 10.1016/j.nbd.2024.106673] [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: 08/06/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited and lethal neurodegenerative disease caused by the abnormal expansion of CAG repeats in the ATAXIN-1 (ATXN1) gene. Pathological studies identified dysfunction and loss of motor neurons (MNs) in the brain stem and spinal cord, which are thought to contribute to premature lethality by affecting the swallowing and breathing of SCA1 patients. However, the molecular and cellular mechanisms of MN pathogenesis remain unknown. To study SCA1 pathogenesis in human MNs, we differentiated induced pluripotent stem cells (iPSCs) derived from SCA1 patients and their unaffected siblings into MNs. We examined proliferation of progenitor cells, neurite outgrowth, spontaneous and glutamate-induced calcium activity of SCA1 MNs to investigate cellular mechanisms of pathogenesis. RNA sequencing was then used to identify transcriptional alterations in iPSC-derived MN progenitors (pMNs) and MNs which could underlie functional changes in SCA1 MNs. We found significantly decreased spontaneous and evoked calcium activity and identified dysregulation of genes regulating calcium signaling in SCA1 MNs. These results indicate that expanded ATXN1 causes dysfunctional calcium signaling in human MNs.
Collapse
Affiliation(s)
- Carrie Sheeler
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America; Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, United States of America
| | - Emmanuel Labrada
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America
| | - Lisa Duvick
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States of America
| | - Leslie M Thompson
- Departments of Psychiatry and Human Behavior and Neurobiology and Behavior, University of California, Irvine, United States of America
| | - Ying Zhang
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America
| | - Harry T Orr
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States of America; Department of Lab Pathology, University of Minnesota, Minneapolis, MN, United States of America
| | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States of America; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States of America.
| |
Collapse
|
2
|
Vohra A, Keefe P, Puthanveetil P. Altered Metabolic Signaling and Potential Therapies in Polyglutamine Diseases. Metabolites 2024; 14:320. [PMID: 38921455 PMCID: PMC11205831 DOI: 10.3390/metabo14060320] [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: 04/30/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
Polyglutamine diseases comprise a cluster of genetic disorders involving neurodegeneration and movement disabilities. In polyglutamine diseases, the target proteins become aberrated due to polyglutamine repeat formation. These aberrant proteins form the root cause of associated complications. The metabolic regulation during polyglutamine diseases is not well studied and needs more attention. We have brought to light the significance of regulating glutamine metabolism during polyglutamine diseases, which could help in decreasing the neuronal damage associated with excess glutamate and nucleotide generation. Most polyglutamine diseases are accompanied by symptoms that occur due to excess glutamate and nucleotide accumulation. Along with a dysregulated glutamine metabolism, the Nicotinamide adenine dinucleotide (NAD+) levels drop down, and, under these conditions, NAD+ supplementation is the only achievable strategy. NAD+ is a major co-factor in the glutamine metabolic pathway, and it helps in maintaining neuronal homeostasis. Thus, strategies to decrease excess glutamate and nucleotide generation, as well as channelizing glutamine toward the generation of ATP and the maintenance of NAD+ homeostasis, could aid in neuronal health. Along with understanding the metabolic dysregulation that occurs during polyglutamine diseases, we have also focused on potential therapeutic strategies that could provide direct benefits or could restore metabolic homeostasis. Our review will shed light into unique metabolic causes and into ideal therapeutic strategies for treating complications associated with polyglutamine diseases.
Collapse
Affiliation(s)
- Alisha Vohra
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (A.V.); (P.K.)
| | - Patrick Keefe
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (A.V.); (P.K.)
| | - Prasanth Puthanveetil
- College of Graduate Studies, Department of Pharmacology, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
3
|
Sucha M, Benediktova S, Tichanek F, Jedlicka J, Kapl S, Jelinkova D, Purkartova Z, Tuma J, Kuncova J, Cendelin J. Experimental Treatment with Edaravone in a Mouse Model of Spinocerebellar Ataxia 1. Int J Mol Sci 2023; 24:10689. [PMID: 37445867 DOI: 10.3390/ijms241310689] [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: 05/31/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Edaravone is a mitochondrially targeted drug with a suggested capability to modify the course of diverse neurological diseases. Nevertheless, edaravone has not been tested yet in the context of spinocerebellar ataxia 1 (SCA1), an incurable neurodegenerative disease characterized mainly by cerebellar disorder, with a strong contribution of inflammation and mitochondrial dysfunction. This study aimed to address this gap, exploring the potential of edaravone to slow down SCA1 progression in a mouse knock-in SCA1 model. SCA1154Q/2Q and healthy SCA12Q/2Q mice were administered either edaravone or saline daily for more than 13 weeks. The functional impairments were assessed via a wide spectrum of behavioral assays reflecting motor and cognitive deficits and behavioral abnormalities. Moreover, we used high-resolution respirometry to explore mitochondrial function, and immunohistochemical and biochemical tools to assess the magnitude of neurodegeneration, inflammation, and neuroplasticity. Data were analyzed using (hierarchical) Bayesian regression models, combined with the methods of multivariate statistics. Our analysis pointed out various previously documented neurological and behavioral deficits of SCA1 mice. However, we did not detect any plausible therapeutic effect of edaravone on either behavioral dysfunctions or other disease hallmarks in SCA1 mice. Thus, our results did not provide support for the therapeutic potential of edaravone in SCA1.
Collapse
Affiliation(s)
- Martina Sucha
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
- Laboratory of Neurodegenerative Disorders, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Simona Benediktova
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Filip Tichanek
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
- Laboratory of Neurodegenerative Disorders, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Jan Jedlicka
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
- Mitochondrial Laboratory, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Stepan Kapl
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
- Laboratory of Experimental Neurophysiology, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Dana Jelinkova
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
- Laboratory of Neurodegenerative Disorders, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Zdenka Purkartova
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Jan Tuma
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Jitka Kuncova
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
- Mitochondrial Laboratory, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Jan Cendelin
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
- Laboratory of Neurodegenerative Disorders, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 76, 323 00 Pilsen, Czech Republic
| |
Collapse
|
4
|
Soles A, Selimovic A, Sbrocco K, Ghannoum F, Hamel K, Moncada EL, Gilliat S, Cvetanovic M. Extracellular Matrix Regulation in Physiology and in Brain Disease. Int J Mol Sci 2023; 24:7049. [PMID: 37108212 PMCID: PMC10138624 DOI: 10.3390/ijms24087049] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The extracellular matrix (ECM) surrounds cells in the brain, providing structural and functional support. Emerging studies demonstrate that the ECM plays important roles during development, in the healthy adult brain, and in brain diseases. The aim of this review is to briefly discuss the physiological roles of the ECM and its contribution to the pathogenesis of brain disease, highlighting the gene expression changes, transcriptional factors involved, and a role for microglia in ECM regulation. Much of the research conducted thus far on disease states has focused on "omic" approaches that reveal differences in gene expression related to the ECM. Here, we review recent findings on alterations in the expression of ECM-associated genes in seizure, neuropathic pain, cerebellar ataxia, and age-related neurodegenerative disorders. Next, we discuss evidence implicating the transcription factor hypoxia-inducible factor 1 (HIF-1) in regulating the expression of ECM genes. HIF-1 is induced in response to hypoxia, and also targets genes involved in ECM remodeling, suggesting that hypoxia could contribute to ECM remodeling in disease conditions. We conclude by discussing the role microglia play in the regulation of the perineuronal nets (PNNs), a specialized form of ECM in the central nervous system. We show evidence that microglia can modulate PNNs in healthy and diseased brain states. Altogether, these findings suggest that ECM regulation is altered in brain disease, and highlight the role of HIF-1 and microglia in ECM remodeling.
Collapse
Affiliation(s)
- Alyssa Soles
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Adem Selimovic
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Kaelin Sbrocco
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Ferris Ghannoum
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Katherine Hamel
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Emmanuel Labrada Moncada
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Stephen Gilliat
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
| | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, 321 Church St SE, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
5
|
Jain M, Patil N, Abdi G, Abbasi Tarighat M, Mohammed A, Ahmad Mohd Zain MR, Goh KW. Mechanistic Insights and Potential Therapeutic Approaches in PolyQ Diseases via Autophagy. Biomedicines 2023; 11:biomedicines11010162. [PMID: 36672670 PMCID: PMC9856063 DOI: 10.3390/biomedicines11010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 01/11/2023] Open
Abstract
Polyglutamine diseases are a group of congenital neurodegenerative diseases categorized with genomic abnormalities in the expansion of CAG triplet repeats in coding regions of specific disease-related genes. Protein aggregates are the toxic hallmark for polyQ diseases and initiate neuronal death. Autophagy is a catabolic process that aids in the removal of damaged organelles or toxic protein aggregates, a process required to maintain cellular homeostasis that has the potential to fight against neurodegenerative diseases, but this pathway gets affected under diseased conditions, as there is a direct impact on autophagy-related gene expression. The increase in the accumulation of autophagy vesicles reported in neurodegenerative diseases was due to an increase in autophagy or may have been due to a decrease in autophagy flux. These reports suggested that there is a contribution of autophagy in the pathology of diseases and regulation in the process of autophagy. It was demonstrated in various disease models of polyQ diseases that autophagy upregulation by using modulators can enhance the dissolution of toxic aggregates and delay disease progression. In this review, interaction of the autophagy pathway with polyQ diseases was analyzed, and a therapeutic approach with autophagy inducing drugs was established for disease pathogenesis.
Collapse
Affiliation(s)
- Mukul Jain
- Department of Lifesciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India
- Lab 209 Cell and Developmental Biology Lab, Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Nil Patil
- Department of Lifesciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India
- Lab 209 Cell and Developmental Biology Lab, Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Gholamreza Abdi
- Department of Biotechnology, Persian Gulf Research Institute, Persian Gulf University, Bushehr, 75169, Iran
- Correspondence: (G.A.); (M.R.A.M.Z.); (K.W.G.)
| | - Maryam Abbasi Tarighat
- Faculty of Nano and Bio Science and Technology, Persian Gulf University, Bushehr 75169, Iran
| | - Arifullah Mohammed
- Department of Agriculture, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Malaysia
| | - Muhammad Rajaei Ahmad Mohd Zain
- Department of Orthopaedics, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Correspondence: (G.A.); (M.R.A.M.Z.); (K.W.G.)
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
- Correspondence: (G.A.); (M.R.A.M.Z.); (K.W.G.)
| |
Collapse
|
6
|
Huang H, Toker N, Burr E, Okoro J, Moog M, Hearing C, Lagalwar S. Intercellular Propagation and Aggregate Seeding of Mutant Ataxin-1. J Mol Neurosci 2021; 72:708-718. [PMID: 34826062 PMCID: PMC8986690 DOI: 10.1007/s12031-021-01944-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/06/2021] [Indexed: 01/07/2023]
Abstract
Intercellular propagation of aggregated protein inclusions along actin-based tunneling nanotubes (TNTs) has been reported as a means of pathogenic spread in Alzheimer’s, Parkinson’s, and Huntington’s diseases. Propagation of oligomeric-structured polyglutamine-expanded ataxin-1 (Atxn1[154Q]) has been reported in the cerebellum of a Spinocerebellar ataxia type 1 (SCA1) knock-in mouse to correlate with disease propagation. In this study, we investigated whether a physiologically relevant polyglutamine-expanded ATXN1 protein (ATXN1[82Q]) could propagate intercellularly. Using a cerebellar-derived live cell model, we observed ATXN1 aggregates form in the nucleus, subsequently form in the cytoplasm, and finally, propagate to neighboring cells along actin-based intercellular connections. Additionally, we observed the facilitation of aggregate-resistant proteins into aggregates given the presence of aggregation-prone proteins within cells. Taken together, our results support a pathogenic role of intercellular propagation of polyglutamine-expanded ATXN1 inclusions.
Collapse
Affiliation(s)
- Haoyang Huang
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Nicholas Toker
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Eliza Burr
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Jeff Okoro
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Maia Moog
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Casey Hearing
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA
| | - Sarita Lagalwar
- Neuroscience Program, Skidmore College, Saratoga Springs, NY, USA.
| |
Collapse
|
7
|
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: 2.0] [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
|
8
|
Gazulla J, Izquierdo-Alvarez S, Ruiz-Fernández E, Berciano J. Initial Cerebellar Ataxia in Hereditary Adult-Onset Primary Lateral Sclerosis. Case Rep Neurol 2021; 13:414-421. [PMID: 34326749 PMCID: PMC8299400 DOI: 10.1159/000515157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
Cerebellar ataxia preceding the apparition of primary lateral sclerosis (PLS) is reported herein. Three individuals from 2 independent kindreds experienced ataxia before developing clinical signs of PLS. Disease onset was during the sixth decade or later, and an insidious onset, with progression exceeding 11 years, was observed. Pathochrony was homogenous, consisting of initial gait instability, followed by hand dysmetria 2 years later. During a 5-year follow-up, cerebellar ataxia remained the sole clinical manifestation, preceding the appearance of muscle stiffness, which progressed to a paraparesis, and then to a purely spastic quadriparesis, over 4 years; pseudobulbar dysarthria and dysphagia appeared later. At this disease stage, limb spasticity, hyperactive jaw and limb stretch reflexes, extensor plantar responses, and a spastic dysarthria were found on examination; limb dysmetria and an ataxo-spastic gait were also found. No muscle atrophy or fasciculation was observed. Among ancillary tests, electromyographic studies performed 6 years after disease onset revealed normal motor unit action potentials and absence of spontaneous activity, in 2 individuals. MRI revealed normal cerebellum and brainstem in 2 cases. Inheritance was dominant in both kindreds, and extensive genetic testing was negative. It is concluded that cerebellar ataxia preceded the appearance of a purely spastic spinobulbar syndrome (which fulfilled the clinical diagnostic criteria for PLS) during a 5-year period in 3 patients with a hereditary, adult-onset form of PLS; subsequent disease progression was equivalent to that of sporadic PLS. Further studies are needed to fully delineate the clinical and genetic spectra of adult-onset PLS.
Collapse
Affiliation(s)
- José Gazulla
- Department of Neurology, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Silvia Izquierdo-Alvarez
- Section of Genetics, Department of Clinical Biochemistry, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | | | - José Berciano
- Department of Neurology, Hospital Universitario Marqués de Valdecilla (IDIVAL), University of Cantabria, CIBERNED, Santander, Spain
| |
Collapse
|
9
|
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by abnormal expansion of glutamine-encoding CAG repeats in the Ataxin-1 (ATXN1) gene. SCA1 is characterized by progressive motor deficits, cognitive decline, and mood changes including anxiety and depression, with longer number of repeats correlating with worse disease outcomes. While mouse models have been very useful in understanding etiology of ataxia and cognitive decline, our understanding of mood symptoms in SCA1 has lagged. It remains unclear whether anxiety or depression stem from an underlying brain pathology or as a consequence of living with an untreatable and lethal disease. To increase our understanding of the etiology of SCA1 mood alterations, we used the elevated-plus maze, sucrose preference and forced swim tests to assess mood in four different mouse lines. We found that SCA1 knock-in mice exhibit increased anxiety that correlated with the length of CAG repeats, supporting the idea that underlying brain pathology contributes to SCA1-like anxiety. Additionally, our results support the concept that increased anxiety is caused by non-cerebellar pathology, as Purkinje cell specific SCA1 transgenic mice exhibit decreased anxiety-like behavior. Regarding the molecular mechanism, partial loss of ATXN1 may play a role in anxiety, based on our results for Atxn1 haploinsufficient and null mice.
Collapse
|
10
|
Jiao S, Wang P, Chen Z, Wang C, Shi Y, Qiu R, Tang B, Jiang H. Age is an important independent modifier of SCA3 phenotype severity. Neurosci Lett 2020; 741:135510. [PMID: 33221475 DOI: 10.1016/j.neulet.2020.135510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/12/2020] [Accepted: 11/15/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE This study aimed to investigate factors modulating spinocerebellar ataxia type 3 (SCA3) phenotype severity besides the expanded CAG repeats (ExpCAG) of ATXN3. METHODS Data regarding CAG trinucleotide repeats, age at onset (AO), duration, age, sex, transmitting parent, and scale scores of SCA3 patients were collected. Multiple linear regression analysis was performed to identify influential independent variables. Age, AO, ExpCAG, and duration were considered control variables to analyze the correlation between independent variables and scale scores. RESULTS Duration, age, and ExpCAG were screened as influential independent variables (P = 0.000). Age had the greatest impact on multiple linear regression models (P<5E-8). ExpCAG and SARA/ICARS/INAS/Barthel index were not correlated (P > 0.05); considering only age as the control, ExpCAG was slightly-to-moderately correlated with all aforementioned scores except INAS (P < 0.05). Age and all scores, except INAS, were positively correlated (P < 0.05); considering duration, AO, or ExpCAG as controls, their correlations did not change significantly. On controlling age, AO was negatively correlated with all scores (P < 0.05), except for the Barthel index (P > 0.05). Furthermore, the interaction model revealed that the interaction between age, duration, and ExpCAG was significantly associated with SCA3 disease severity (P < 0.05). CONCLUSION Age is a potentially important modifier of SCA3 phenotype severity, through the interaction between ExpCAG and aging factors.
Collapse
Affiliation(s)
- Shujun Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Puzhi Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Diseases, Central South University, Changsha, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Chunrong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuting Shi
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Rong Qiu
- School of Information Science and Engineering, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Diseases, Central South University, Changsha, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China; Laboratory of Medical Genetics, Central South University, Changsha, China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Diseases, Central South University, Changsha, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China; Laboratory of Medical Genetics, Central South University, Changsha, China.
| |
Collapse
|
11
|
Neurochemical Differences in Spinocerebellar Ataxia Type 14 and 1. THE CEREBELLUM 2020; 20:169-178. [PMID: 33063293 PMCID: PMC8004522 DOI: 10.1007/s12311-020-01201-y] [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] [Accepted: 10/04/2020] [Indexed: 11/25/2022]
Abstract
Autosomal-dominant spinocerebellar ataxias (SCA) are neurodegenerative diseases characterized by progressive ataxia. Here, we report on neurometabolic alterations in spinocerebellar ataxia type 1 (SCA1; SCA-ATXN1) and 14 (SCA14; SCA-PRKCG) assessed by non-invasive 1H magnetic resonance spectroscopy. Three Tesla 1H magnetic resonance spectroscopy was performed in 17 SCA14, 14 SCA1 patients, and in 31 healthy volunteers. We assessed metabolites in the cerebellar vermis, right cerebellar hemisphere, pons, prefrontal, and motor cortex. Additionally, clinical characteristics were obtained for each patient to correlate them with metabolites. In SCA14, metabolic changes were restricted to the cerebellar vermis compared with widespread neurochemical alterations in SCA1. In SCA14, total N-acetylaspartate (tNAA) was reduced in the vermis by 34%. In SCA1, tNAA was reduced in the vermis (24%), cerebellar hemisphere (26%), and pons (25%). SCA14 patients showed 24% lower glutamate+glutamine (Glx) and 46% lower γ-aminobutyric acid (GABA) in the vermis, while SCA1 patients showed no alterations in Glx and GABA. SCA1 revealed a decrease of aspartate (Asp) in the vermis (62%) and an elevation in the prefrontal cortex (130%) as well as an elevation of myo-inositol (Ins) in the cerebellar hemisphere (51%) and pons (46%). No changes of Asp and Ins were detected in SCA14. Beyond, glucose (Glc) was increased in the vermis of both SCA14 (155%) and SCA1 (247%). 1H magnetic resonance spectroscopy revealed differing neurochemical profiles in SCA1 and SCA14 and confirmed metabolic changes that may be indicative for neuronal loss and dysfunctional energy metabolism. Therefore, 1H magnetic resonance spectroscopy represents a helpful tool for in-vivo tracking of disease-specific pathophysiology.
Collapse
|
12
|
Owada R, Awata S, Suzue K, Kanetaka H, Kakuta Y, Nakamura K. Polyglutamine-containing microglia leads to disturbed differentiation and neurite retraction of neuron-like cells. Heliyon 2020; 6:e04851. [PMID: 32954034 PMCID: PMC7486442 DOI: 10.1016/j.heliyon.2020.e04851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 04/13/2020] [Accepted: 09/02/2020] [Indexed: 11/17/2022] Open
Abstract
Expanded polyglutamine-containing proteins in neurons intrinsically contributes to neuronal dysfunctions and neuronal cell death in polyglutamine (polyQ) diseases. In addition, an expanded polyQ-containing protein in microglia also leads to apoptosis of neurons. However, detailed morphological analysis of neurons exposed to conditioned medium (CM) derived from polyQ-containing microglia has not been essentially carried out. Here, we introduced aggregated peptide with 69 glutamine repeat (69Q) into BV2 microglial cells. The 69Q-containing BV2 cells showed shorter branches. The CM from 69Q-containing microglia (69Q-CM) induced neurite retraction and fewer number of branch point of neurites of differentiated PC12 cells. Likewise, the 69Q-CM induces disturbed differentiation of PC12 cells with shorter total length of neurites and fewer number of branch point of neurites. Thus, the factor(s) released from polyQ-containing microglia affect both differentiation and degeneration of neuron-like cells.
Collapse
Affiliation(s)
- Ryuji Owada
- Gunma University Graduate School of Health Sciences, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Saaya Awata
- Gunma University Graduate School of Health Sciences, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Kazutomo Suzue
- Department of Parasitology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hiroyasu Kanetaka
- Laison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Yohei Kakuta
- Department of Orthopedic Surgery, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
- Corresponding author.
| | - Kazuhiro Nakamura
- Gunma University Graduate School of Health Sciences, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
- Corresponding author.
| |
Collapse
|
13
|
Tichanek F, Salomova M, Jedlicka J, Kuncova J, Pitule P, Macanova T, Petrankova Z, Tuma Z, Cendelin J. Hippocampal mitochondrial dysfunction and psychiatric-relevant behavioral deficits in spinocerebellar ataxia 1 mouse model. Sci Rep 2020; 10:5418. [PMID: 32214165 PMCID: PMC7096488 DOI: 10.1038/s41598-020-62308-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
Spinocerebellar ataxia 1 (SCA1) is a devastating neurodegenerative disease associated with cerebellar degeneration and motor deficits. However, many patients also exhibit neuropsychiatric impairments such as depression and apathy; nevertheless, the existence of a causal link between the psychiatric symptoms and SCA1 neuropathology remains controversial. This study aimed to explore behavioral deficits in a knock-in mouse SCA1 (SCA1154Q/2Q) model and to identify the underlying neuropathology. We found that the SCA1 mice exhibit previously undescribed behavioral impairments such as increased anxiety- and depressive-like behavior and reduced prepulse inhibition and cognitive flexibility. Surprisingly, non-motor deficits characterize the early SCA1 stage in mice better than does ataxia. Moreover, the SCA1 mice exhibit significant hippocampal atrophy with decreased plasticity-related markers and markedly impaired neurogenesis. Interestingly, the hippocampal atrophy commences earlier than the cerebellar degeneration and directly reflects the individual severity of some of the behavioral deficits. Finally, mitochondrial respirometry suggests profound mitochondrial dysfunction in the hippocampus, but not in the cerebellum of the young SCA1 mice. These findings imply the essential role of hippocampal impairments, associated with profound mitochondrial dysfunction, in SCA1 behavioral deficits. Moreover, they underline the view of SCA1 as a complex neurodegenerative disease and suggest new avenues in the search for novel SCA1 therapies.
Collapse
Affiliation(s)
- Filip Tichanek
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia. .,Laboratory of Neurodegenerative Disorders, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.
| | - Martina Salomova
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Laboratory of Neurodegenerative Disorders, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Jan Jedlicka
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Mitochondrial Laboratory, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Jitka Kuncova
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Mitochondrial Laboratory, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Pavel Pitule
- Laboratory of Tumor Biology, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Tereza Macanova
- Department of Biology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Zuzana Petrankova
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Zdenek Tuma
- Laboratory of Proteomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Jan Cendelin
- Department of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia.,Laboratory of Neurodegenerative Disorders, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| |
Collapse
|
14
|
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]
|
15
|
Prestori F, Moccia F, D’Angelo E. Disrupted Calcium Signaling in Animal Models of Human Spinocerebellar Ataxia (SCA). Int J Mol Sci 2019; 21:ijms21010216. [PMID: 31892274 PMCID: PMC6981692 DOI: 10.3390/ijms21010216] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/22/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
Spinocerebellar ataxias (SCAs) constitute a heterogeneous group of more than 40 autosomal-dominant genetic and neurodegenerative diseases characterized by loss of balance and motor coordination due to dysfunction of the cerebellum and its efferent connections. Despite a well-described clinical and pathological phenotype, the molecular and cellular events that underlie neurodegeneration are still poorly undaerstood. Emerging research suggests that mutations in SCA genes cause disruptions in multiple cellular pathways but the characteristic SCA pathogenesis does not begin until calcium signaling pathways are disrupted in cerebellar Purkinje cells. Ca2+ signaling in Purkinje cells is important for normal cellular function as these neurons express a variety of Ca2+ channels, Ca2+-dependent kinases and phosphatases, and Ca2+-binding proteins to tightly maintain Ca2+ homeostasis and regulate physiological Ca2+-dependent processes. Abnormal Ca2+ levels can activate toxic cascades leading to characteristic death of Purkinje cells, cerebellar atrophy, and ataxia that occur in many SCAs. The output of the cerebellar cortex is conveyed to the deep cerebellar nuclei (DCN) by Purkinje cells via inhibitory signals; thus, Purkinje cell dysfunction or degeneration would partially or completely impair the cerebellar output in SCAs. In the absence of the inhibitory signal emanating from Purkinje cells, DCN will become more excitable, thereby affecting the motor areas receiving DCN input and resulting in uncoordinated movements. An outstanding advantage in studying the pathogenesis of SCAs is represented by the availability of a large number of animal models which mimic the phenotype observed in humans. By mainly focusing on mouse models displaying mutations or deletions in genes which encode for Ca2+ signaling-related proteins, in this review we will discuss the several pathogenic mechanisms related to deranged Ca2+ homeostasis that leads to significant Purkinje cell degeneration and dysfunction.
Collapse
Affiliation(s)
- Francesca Prestori
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
- Correspondence:
| | - Francesco Moccia
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy;
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| |
Collapse
|
16
|
Naphade S, Tshilenge KT, Ellerby LM. Modeling Polyglutamine Expansion Diseases with Induced Pluripotent Stem Cells. Neurotherapeutics 2019; 16:979-998. [PMID: 31792895 PMCID: PMC6985408 DOI: 10.1007/s13311-019-00810-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Polyglutamine expansion disorders, which include Huntington's disease, have expanded CAG repeats that result in polyglutamine expansions in affected proteins. How this specific feature leads to distinct neuropathies in 11 different diseases is a fascinating area of investigation. Most proteins affected by polyglutamine expansions are ubiquitously expressed, yet their mechanisms of selective neurotoxicity are unknown. Induced pluripotent stem cells have emerged as a valuable tool to model diseases, understand molecular mechanisms, and generate relevant human neural and glia subtypes, cocultures, and organoids. Ideally, this tool will generate specific neuronal populations that faithfully recapitulate specific polyglutamine expansion disorder phenotypes and mimic the selective vulnerability of a given disease. Here, we review how induced pluripotent technology is used to understand the effects of the disease-causing polyglutamine protein on cell function, identify new therapeutic targets, and determine how polyglutamine expansion affects human neurodevelopment and disease. We will discuss ongoing challenges and limitations in our use of induced pluripotent stem cells to model polyglutamine expansion diseases.
Collapse
Affiliation(s)
- Swati Naphade
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA
| | | | - Lisa M Ellerby
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| |
Collapse
|
17
|
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
|
18
|
Cobos SN, Bennett SA, Torrente MP. The impact of histone post-translational modifications in neurodegenerative diseases. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1982-1991. [PMID: 30352259 PMCID: PMC6475498 DOI: 10.1016/j.bbadis.2018.10.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/05/2018] [Accepted: 10/11/2018] [Indexed: 02/08/2023]
Abstract
Every year, neurodegenerative disorders take more than 5000 lives in the US alone. Cures have not yet been found for many of the multitude of neuropathies. The majority of amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD) and Parkinson's disease (PD) cases have no known genetic basis. Thus, it is evident that contemporary genetic approaches have failed to explain the etiology or etiologies of ALS/FTD and PD. Recent investigations have explored the potential role of epigenetic mechanisms in disease development. Epigenetics comprises heritable changes in gene utilization that are not derived from changes in the genome. A main epigenetic mechanism involves the post-translational modification of histones. Increased knowledge of the epigenomic landscape of neurodegenerative diseases would not only further our understanding of the disease pathologies, but also lead to the development of treatments able to halt their progress. Here, we review recent advances on the association of histone post-translational modifications with ALS, FTD, PD and several ataxias.
Collapse
Affiliation(s)
- Samantha N Cobos
- Chemistry Department of Brooklyn College, Brooklyn, New York 11210, United States; Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States
| | - Seth A Bennett
- Chemistry Department of Brooklyn College, Brooklyn, New York 11210, United States; Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States
| | - Mariana P Torrente
- Chemistry Department of Brooklyn College, Brooklyn, New York 11210, United States; Ph.D. Programs in Chemistry, Biochemistry, and Biology, The Graduate Center of the City University of New York, New York 10016, United States.
| |
Collapse
|
19
|
Buijsen RAM, Toonen LJA, Gardiner SL, van Roon-Mom WMC. Genetics, Mechanisms, and Therapeutic Progress in Polyglutamine Spinocerebellar Ataxias. Neurotherapeutics 2019; 16:263-286. [PMID: 30607747 PMCID: PMC6554265 DOI: 10.1007/s13311-018-00696-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Autosomal dominant cerebellar ataxias (ADCAs) are a group of neurodegenerative disorders characterized by degeneration of the cerebellum and its connections. All ADCAs have progressive ataxia as their main clinical feature, frequently accompanied by dysarthria and oculomotor deficits. The most common spinocerebellar ataxias (SCAs) are 6 polyglutamine (polyQ) SCAs. These diseases are all caused by a CAG repeat expansion in the coding region of a gene. Currently, no curative treatment is available for any of the polyQ SCAs, but increasing knowledge on the genetics and the pathological mechanisms of these polyQ SCAs has provided promising therapeutic targets to potentially slow disease progression. Potential treatments can be divided into pharmacological and gene therapies that target the toxic downstream effects, gene therapies that target the polyQ SCA genes, and stem cell replacement therapies. Here, we will provide a review on the genetics, mechanisms, and therapeutic progress in polyglutamine spinocerebellar ataxias.
Collapse
Affiliation(s)
- Ronald A M Buijsen
- Department of Human Genetics, LUMC, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Lodewijk J A Toonen
- Department of Human Genetics, LUMC, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Sarah L Gardiner
- Department of Human Genetics, LUMC, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
- Department of Neurology, LUMC, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | | |
Collapse
|
20
|
Gazulla J, Ferrer I, Izquierdo-Alvarez S, Alvarez S, Sánchez-Alcudia R, Bestué-Cardiel M, Seral M, Benavente I, Sierra-Martínez E, Berciano J. Hereditary primary lateral sclerosis and progressive nonfluent aphasia. J Neurol 2019; 266:1079-1090. [DOI: 10.1007/s00415-019-09235-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 12/31/2022]
|
21
|
François-Moutal L, Jahanbakhsh S, Nelson ADL, Ray D, Scott DD, Hennefarth MR, Moutal A, Perez-Miller S, Ambrose AJ, Al-Shamari A, Coursodon P, Meechoovet B, Reiman R, Lyons E, Beilstein M, Chapman E, Morris QD, Van Keuren-Jensen K, Hughes TR, Khanna R, Koehler C, Jen J, Gokhale V, Khanna M. A Chemical Biology Approach to Model Pontocerebellar Hypoplasia Type 1B (PCH1B). ACS Chem Biol 2018; 13:3000-3010. [PMID: 30141626 DOI: 10.1021/acschembio.8b00745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mutations of EXOSC3 have been linked to the rare neurological disorder known as Pontocerebellar Hypoplasia type 1B (PCH1B). EXOSC3 is one of three putative RNA-binding structural cap proteins that guide RNA into the RNA exosome, the cellular machinery that degrades RNA. Using RNAcompete, we identified a G-rich RNA motif binding to EXOSC3. Surface plasmon resonance (SPR) and microscale thermophoresis (MST) indicated an affinity in the low micromolar range of EXOSC3 for long and short G-rich RNA sequences. Although several PCH1B-causing mutations in EXOSC3 did not engage a specific RNA motif as shown by RNAcompete, they exhibited lower binding affinity to G-rich RNA as demonstrated by MST. To test the hypothesis that modification of the RNA-protein interface in EXOSC3 mutants may be phenocopied by small molecules, we performed an in-silico screen of 50 000 small molecules and used enzyme-linked immunosorbant assays (ELISAs) and MST to assess the ability of the molecules to inhibit RNA-binding by EXOSC3. We identified a small molecule, EXOSC3-RNA disrupting (ERD) compound 3 (ERD03), which ( i) bound specifically to EXOSC3 in saturation transfer difference nuclear magnetic resonance (STD-NMR), ( ii) disrupted the EXOSC3-RNA interaction in a concentration-dependent manner, and ( iii) produced a PCH1B-like phenotype with a 50% reduction in the cerebellum and an abnormally curved spine in zebrafish embryos. This compound also induced modification of zebrafish RNA expression levels similar to that observed with a morpholino against EXOSC3. To our knowledge, this is the first example of a small molecule obtained by rational design that models the abnormal developmental effects of a neurodegenerative disease in a whole organism.
Collapse
Affiliation(s)
- Liberty François-Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
- Center for Innovation in Brain Science, Tucson, Arizona 85721, United States
| | - Shahriyar Jahanbakhsh
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Andrew D. L. Nelson
- School of Plant Sciences, University of Arizona, Tucson, Arizona 85721, United States
| | - Debashish Ray
- Donnelly Centre, University of Toronto, Toronto, Canada M5S 3E1
| | - David D. Scott
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
- Center for Innovation in Brain Science, Tucson, Arizona 85721, United States
| | - Matthew R. Hennefarth
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
- Center for Innovation in Brain Science, Tucson, Arizona 85721, United States
| | - Andrew J. Ambrose
- Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona 85724, United States
| | - Ahmed Al-Shamari
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | - Philippe Coursodon
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
| | | | - Rebecca Reiman
- Neurogenomics Division, TGen, Phoenix, Arizona 85004, United States
| | - Eric Lyons
- School of Plant Sciences, University of Arizona, Tucson, Arizona 85721, United States
| | - Mark Beilstein
- School of Plant Sciences, University of Arizona, Tucson, Arizona 85721, United States
| | - Eli Chapman
- Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona 85724, United States
| | - Quaid D. Morris
- Donnelly Centre, University of Toronto, Toronto, Canada M5S 3E1
- Department of Molecular Genetics, University of Toronto, Toronto, Canada M5S 1A8
- Department of Computer Science, University of Toronto, Toronto, Canada M5S 2E4
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, Canada M5S3G4
| | | | - Timothy R. Hughes
- Donnelly Centre, University of Toronto, Toronto, Canada M5S 3E1
- Department of Molecular Genetics, University of Toronto, Toronto, Canada M5S 1A8
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
- Center for Innovation in Brain Science, Tucson, Arizona 85721, United States
| | - Carla Koehler
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Joanna Jen
- Mount Sinai, New York, New York 10029, United States
| | - Vijay Gokhale
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States
| | - May Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States
- Center for Innovation in Brain Science, Tucson, Arizona 85721, United States
| |
Collapse
|
22
|
Pilotto F, Saxena S. Epidemiology of inherited cerebellar ataxias and challenges in clinical research. CLINICAL AND TRANSLATIONAL NEUROSCIENCE 2018. [DOI: 10.1177/2514183x18785258] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Federica Pilotto
- Department of Neurology, Inselspital University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Regenerative Neuroscience Cluster, University of Bern, Bern, Switzerland
| | - Smita Saxena
- Department of Neurology, Inselspital University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Regenerative Neuroscience Cluster, University of Bern, Bern, Switzerland
| |
Collapse
|
23
|
Kim JH, Lukowicz A, Qu W, Johnson A, Cvetanovic M. Astroglia contribute to the pathogenesis of spinocerebellar ataxia Type 1 (SCA1) in a biphasic, stage-of-disease specific manner. Glia 2018; 66:1972-1987. [PMID: 30043530 DOI: 10.1002/glia.23451] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/11/2018] [Accepted: 04/11/2018] [Indexed: 12/22/2022]
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a fatal, dominantly inherited neurodegenerative disease caused by the expansion of CAG repeats in the Ataxin-1 (ATXN1) gene. SCA1 is characterized by balance and coordination deficits due to the predominant loss of Purkinje neurons in the cerebellum. We previously demonstrated that cerebellar astrogliosis beings during the early stages of SCA1, prior to onset of motor deficits and loss of Purkinje neurons. We communicate here that cerebellar astrogliosis contributes to SCA1 pathogenesis in a biphasic, stage of disease dependent manner. We modulated astrogliosis by selectively reducing pro-inflammatory transcriptional regulator nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) signaling in astroglia via a Cre-lox mouse genetic approach. Our results indicate that inhibition of astroglial NF-κB signaling, prior to motor deficit onset, exacerbates disease severity. This is suggestive of a neuroprotective role mediated by astroglia during early stage SCA1. In contrast, inhibition of astroglial NF-κB signaling during late stage of disease ameliorated motor deficits, indicating a potentially harmful role of astroglia late in SCA1. These results indicate that astrogliosis may have a critical and dual role in disease. If so, our results imply that anti-inflammatory astroglia-based therapeutic approaches may need to consider disease progression to achieve therapeutic efficacy.
Collapse
Affiliation(s)
- Joo Hyun Kim
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota 2101 6th Street SE, Minneapolis, Minnesota
| | - Abigail Lukowicz
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota 2101 6th Street SE, Minneapolis, Minnesota
| | - Wenhui Qu
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota 2101 6th Street SE, Minneapolis, Minnesota
| | - Andrea Johnson
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota 2101 6th Street SE, Minneapolis, Minnesota
| | - Marija Cvetanovic
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota 2101 6th Street SE, Minneapolis, Minnesota
| |
Collapse
|
24
|
Ferro A, Qu W, Lukowicz A, Svedberg D, Johnson A, Cvetanovic M. Inhibition of NF-κB signaling in IKKβF/F;LysM Cre mice causes motor deficits but does not alter pathogenesis of Spinocerebellar ataxia type 1. PLoS One 2018; 13:e0200013. [PMID: 29975753 PMCID: PMC6033432 DOI: 10.1371/journal.pone.0200013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/18/2018] [Indexed: 12/21/2022] Open
Abstract
Spinocerebellar Ataxia type 1 (SCA1) is a fatal neurodegenerative genetic disease that is characterized by pronounced neuronal loss and gliosis in the cerebellum. We have previously demonstrated microglial activation, measured as an increase in microglial density in cerebellar cortex and an increase in the production of pro-inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), in the cerebellum of the ATXN1[82Q] transgenic mouse model of SCA1. To examine the role of activated state of microglia in SCA1, we used a Cre-Lox approach with IKKβF/F;LysM Cre mice intended to reduce inflammatory NF-κB signaling, selectively in microglia. ATXN1[82Q];IKKβF/F;LysM Cre mice showed reduced cerebellar microglial density and production of TNFα compared to ATXN1[82Q] mice, yet reducing NF-κB did not ameliorate motor impairments and cerebellar cellular pathologies. Unexpectedly, at 12 weeks of age, control IKKβF/F;LysM Cre mice showed motor deficits equal to ATXN1[82Q] mice that were dissociated from any obvious neurodegenerative changes in the cerebellum, but were rather associated with a developmental impairment that presented as a retention of climbing fiber synaptic terminals on the soma of Purkinje neurons. These results indicate that NF-κB signaling is required for increase in microglial numbers and TNF-α production in the cerebella of ATXN1[82Q] mouse model of SCA1. Furthermore, these results elucidate a novel role of canonical NF-κB signaling in pruning of surplus synapses on Purkinje neurons in the cerebellum during development.
Collapse
Affiliation(s)
- Austin Ferro
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wenhui Qu
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Abigail Lukowicz
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Daniel Svedberg
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Andrea Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
25
|
Pérez Ortiz JM, Mollema N, Toker N, Adamski CJ, O'Callaghan B, Duvick L, Friedrich J, Walters MA, Strasser J, Hawkinson JE, Zoghbi HY, Henzler C, Orr HT, Lagalwar S. Reduction of protein kinase A-mediated phosphorylation of ATXN1-S776 in Purkinje cells delays onset of Ataxia in a SCA1 mouse model. Neurobiol Dis 2018; 116:93-105. [PMID: 29758256 DOI: 10.1016/j.nbd.2018.05.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/17/2018] [Accepted: 05/09/2018] [Indexed: 12/27/2022] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a polyglutamine (polyQ) repeat neurodegenerative disease in which a primary site of pathogenesis are cerebellar Purkinje cells. In addition to polyQ expansion of ataxin-1 protein (ATXN1), phosphorylation of ATXN1 at the serine 776 residue (ATXN1-pS776) plays a significant role in protein toxicity. Utilizing a biochemical approach, pharmacological agents and cell-based assays, including SCA1 patient iPSC-derived neurons, we examine the role of Protein Kinase A (PKA) as an effector of ATXN1-S776 phosphorylation. We further examine the implications of PKA-mediated phosphorylation at ATXN1-S776 on SCA1 through genetic manipulation of the PKA catalytic subunit Cα in Pcp2-ATXN1[82Q] mice. Here we show that pharmacologic inhibition of S776 phosphorylation in transfected cells and SCA1 patient iPSC-derived neuronal cells lead to a decrease in ATXN1. In vivo, reduction of PKA-mediated ATXN1-pS776 results in enhanced degradation of ATXN1 and improved cerebellar-dependent motor performance. These results provide evidence that PKA is a biologically important kinase for ATXN1-pS776 in cerebellar Purkinje cells.
Collapse
Affiliation(s)
- Judit M Pérez Ortiz
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Nissa Mollema
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Nicholas Toker
- Skidmore College Neuroscience Program, Saratoga Springs, NY, United States
| | - Carolyn J Adamski
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, and Department of Molecular and Human Genetics, Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, United States
| | - Brennon O'Callaghan
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Lisa Duvick
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Jillian Friedrich
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Michael A Walters
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, United States
| | - Jessica Strasser
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, United States
| | - Jon E Hawkinson
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, United States
| | - Huda Y Zoghbi
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, and Department of Molecular and Human Genetics, Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, United States
| | - Christine Henzler
- RISS Bioinformatics, Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, United States
| | - Harry T Orr
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States.
| | - Sarita Lagalwar
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States; Skidmore College Neuroscience Program, Saratoga Springs, NY, United States.
| |
Collapse
|
26
|
Conformation Polymorphism of Polyglutamine Proteins. Trends Biochem Sci 2018; 43:424-435. [PMID: 29636213 DOI: 10.1016/j.tibs.2018.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/05/2018] [Accepted: 03/12/2018] [Indexed: 01/29/2023]
Abstract
Expanded polyglutamine (polyQ) stretches within endogenous proteins cause at least nine human diseases. The structural basis of polyQ pathogenesis is the key to understanding fundamental mechanisms of these diseases, but it remains unclear and controversial due to a lack of polyQ protein structures at the single-atom level. Various hypotheses have been proposed to explain the structure-cytotoxicity relationship of pathogenic proteins with polyQ expansion, largely based on indirect evidence. Here we review these hypotheses and their supporting evidence, along with additional insights from recent structural biology and chemical biology studies, with a focus on Huntingtin (HTT), the most extensively studied polyQ disease protein. Lastly, we propose potential novel strategies that may further clarify the conformation-cytotoxicity relationship of polyQ proteins.
Collapse
|
27
|
Patron M, Sprenger HG, Langer T. m-AAA proteases, mitochondrial calcium homeostasis and neurodegeneration. Cell Res 2018; 28:296-306. [PMID: 29451229 PMCID: PMC5835776 DOI: 10.1038/cr.2018.17] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The function of mitochondria depends on ubiquitously expressed and evolutionary conserved m-AAA proteases in the inner membrane. These ATP-dependent peptidases form hexameric complexes built up of homologous subunits. AFG3L2 subunits assemble either into homo-oligomeric isoenzymes or with SPG7 (paraplegin) subunits into hetero-oligomeric proteolytic complexes. Mutations in AFG3L2 are associated with dominant spinocerebellar ataxia (SCA28) characterized by the loss of Purkinje cells, whereas mutations in SPG7 cause a recessive form of hereditary spastic paraplegia (HSP7) with motor neurons of the cortico-spinal tract being predominantly affected. Pleiotropic functions have been assigned to m-AAA proteases, which act as quality control and regulatory enzymes in mitochondria. Loss of m-AAA proteases affects mitochondrial protein synthesis and respiration and leads to mitochondrial fragmentation and deficiencies in the axonal transport of mitochondria. Moreover m-AAA proteases regulate the assembly of the mitochondrial calcium uniporter (MCU) complex. Impaired degradation of the MCU subunit EMRE in AFG3L2-deficient mitochondria results in the formation of deregulated MCU complexes, increased mitochondrial calcium uptake and increased vulnerability of neurons for calcium-induced cell death. A reduction of calcium influx into the cytosol of Purkinje cells rescues ataxia in an AFG3L2-deficient mouse model. In this review, we discuss the relationship between the m-AAA protease and mitochondrial calcium homeostasis and its relevance for neurodegeneration and describe a novel mouse model lacking MCU specifically in Purkinje cells. Our results pledge for a novel view on m-AAA proteases that integrates their pleiotropic functions in mitochondria to explain the pathogenesis of associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Maria Patron
- Max Planck Institute for Biology of Aging, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Disease (CECAD), and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Hans-Georg Sprenger
- Max Planck Institute for Biology of Aging, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Disease (CECAD), and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Thomas Langer
- Max Planck Institute for Biology of Aging, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Disease (CECAD), and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
28
|
Abstract
Spinocerebellar ataxia type 1 (SCA1) is an autosomal dominant disorder caused by a CAG repeat expansion, characterized by progressive cerebellar ataxia and pyramidal signs. Non-motor and extracerebellar symptoms may occur. MRI-based studies in SCA1 focused in the cerebellum and connections, but there are no data about cord damage in the disease and its clinical relevance. To evaluate in vivo spinal cord damage in SCA1, a group of 31 patients with SCA1 and 31 age- and gender-matched healthy controls underwent MRI on a 3T scanner. We used T1-weighted 3D images to estimate the cervical spinal cord area (CA) and eccentricity (CE) at three C2/C3 levels based on a semi-automatic image segmentation protocol. The scale for assessment and rating of ataxia (SARA) was used to quantify disease severity. The groups were significantly different regarding CA (47.26 ± 7.4 vs. 68.8 ± 5.7 mm2, p < 0.001) and CE values (0.803 ± 0.044 vs. 0.774 ± 0.043, p < 0.05). Furthermore, in the patient group, CA presented significant correlation with SARA scores (R = -0.633, p < 0.001) and CAGn expansion (R = -0.658, p < 0.001). CE was not associated with SARA scores (p = 0.431). In the multiple variable regression, CA was strongly associated with disease duration (coefficient -0.360, p < 0.05) and CAGn expansion (coefficient -1.124, p < 0.001). SCA1 is characterized by cervical cord atrophy and anteroposterior flattening. Morphometric analyses of the spinal cord MRI might be a useful biomarker in the disease.
Collapse
|
29
|
Qu W, Johnson A, Kim JH, Lukowicz A, Svedberg D, Cvetanovic M. Inhibition of colony-stimulating factor 1 receptor early in disease ameliorates motor deficits in SCA1 mice. J Neuroinflammation 2017; 14:107. [PMID: 28545543 PMCID: PMC5445366 DOI: 10.1186/s12974-017-0880-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/12/2017] [Indexed: 12/16/2022] Open
Abstract
Background Polyglutamine (polyQ) expansion in the protein Ataxin-1 (ATXN1) causes spinocerebellar ataxia type 1 (SCA1), a fatal dominantly inherited neurodegenerative disease characterized by motor deficits, cerebellar neurodegeneration, and gliosis. Currently, there are no treatments available to delay or ameliorate SCA1. We have examined the effect of depleting microglia during the early stage of disease by using PLX, an inhibitor of colony-stimulating factor 1 receptor (CSFR1), on disease severity in a mouse model of SCA1. Methods Transgenic mouse model of SCA1, ATXN1[82Q] mice, and wild-type littermate controls were treated with PLX from 3 weeks of age. The effects of PLX on microglial density, astrogliosis, motor behavior, atrophy, and gene expression of Purkinje neurons were examined at 3 months of age. Results PLX treatment resulted in the elimination of 70–80% of microglia from the cerebellum of both wild-type and ATXN1[82Q] mice. Importantly, PLX ameliorated motor deficits in SCA1 mice. While we have not observed significant improvement in the atrophy or disease-associated gene expression changes in Purkinje neurons upon PLX treatment, we have detected reduced expression of pro-inflammatory cytokine tumor necrosis factor alpha (TNFα) and increase in the protein levels of wild-type ataxin-1 and post-synaptic density protein 95 (PSD95) that may help improve PN function. Conclusions A decrease in the number of microglia during an early stage of disease resulted in the amelioration of motor deficits in SCA1 mice. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0880-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenhui Qu
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Andrea Johnson
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Joo Hyun Kim
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Abigail Lukowicz
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Daniel Svedberg
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Marija Cvetanovic
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
30
|
Sánchez I, Balagué E, Matilla-Dueñas A. Ataxin-1 regulates the cerebellar bioenergetics proteome through the GSK3β-mTOR pathway which is altered in Spinocerebellar ataxia type 1 (SCA1). Hum Mol Genet 2016; 25:4021-4040. [PMID: 27466200 DOI: 10.1093/hmg/ddw242] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/21/2016] [Accepted: 07/11/2016] [Indexed: 12/17/2022] Open
Abstract
A polyglutamine expansion within the ataxin-1 protein (ATXN1) underlies spinocerebellar ataxia type-1 (SCA1), a neurological disorder mainly characterized by ataxia and cerebellar deficits. In SCA1, both loss and gain of ATXN1 biological functions contribute to cerebellar pathogenesis. However, the critical ATXN1 functions and pathways involved remain unclear. To further investigate the early signalling pathways regulated by ATXN1, we performed an unbiased proteomic study of the Atxn1-KO 5-week-old mice cerebellum. Here, we show that lack of ATXN1 expression induces early alterations in proteins involved in glycolysis [pyruvate kinase, muscle, isoform 1 protein (PKM-i1), citrate synthase (CS), glycerol-3-phosphate dehydrogenase 2 (GPD2), glucose-6-phosphate isomerase (GPI), alpha -: enolase (ENO1)], ATP synthesis [CS, Succinate dehydrogenase complex,subunit A (SDHA), ATP synthase subunit d, mitochondrial (ATP5H)] and oxidative stress [peroxiredoxin-6 (PRDX6), aldehyde dehydrogenase family 1, subfamily A1, 10-formyltetrahydrofolate dehydrogenase]. In the SCA1 mice, several of these proteins (PKM-i1, ATP5H, PRDX6, proteome subunit A6) were down-regulated and ATP levels decreased. The underlying mechanism does not involve modulation of mitochondrial biogenesis, but dysregulation of the activity of the metabolic regulators glycogen synthase kinase 3B (GSK3β), decreased in Atxn1-KO and increased in SCA1 mice, and mechanistic target of rapamycin (serine/threonine kinase) (mTOR), unchanged in the Atxn1-KO and decreased in SCA1 mice cerebellum before the onset of ataxic symptoms. Pharmacological inhibition of GSK3β and activation of mTOR in a SCA1 cell model ameliorated identified ATXN1-regulated metabolic proteome and ATP alterations. Taken together, these results point to an early role of ATXN1 in the regulation of bioenergetics homeostasis in the mouse cerebellum. Moreover, data suggest GSK3β and mTOR pathways modulate this ATXN1 function in SCA1 pathogenesis that could be targeted therapeutically prior to the onset of disease symptoms in SCA1 and other pathologies involving dysregulation of ATXN1 functions.
Collapse
Affiliation(s)
- Ivelisse Sánchez
- Functional and Translational Neurogenetics Unit, Department of Neurosciences, Health Sciences Research Institute Germans Trias i Pujol (IGTP)-Universitat Autonoma de Barcelona, Crta. de Can Ruti, camí de les escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Eudald Balagué
- Functional and Translational Neurogenetics Unit, Department of Neurosciences, Health Sciences Research Institute Germans Trias i Pujol (IGTP)-Universitat Autonoma de Barcelona, Crta. de Can Ruti, camí de les escoles s/n, 08916 Badalona, Barcelona, Spain
| | - Antoni Matilla-Dueñas
- Functional and Translational Neurogenetics Unit, Department of Neurosciences, Health Sciences Research Institute Germans Trias i Pujol (IGTP)-Universitat Autonoma de Barcelona, Crta. de Can Ruti, camí de les escoles s/n, 08916 Badalona, Barcelona, Spain
| |
Collapse
|
31
|
Giunta M, Edvardson S, Xu Y, Schuelke M, Gomez-Duran A, Boczonadi V, Elpeleg O, Müller JS, Horvath R. Altered RNA metabolism due to a homozygous RBM7 mutation in a patient with spinal motor neuropathy. Hum Mol Genet 2016; 25:2985-2996. [PMID: 27193168 PMCID: PMC5181591 DOI: 10.1093/hmg/ddw149] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 12/23/2022] Open
Abstract
The exosome complex is the most important RNA processing machinery within the cell. Mutations in its subunits EXOSC8 and EXOSC3 cause pontocerebellar hypoplasia, spinal muscular atrophy (SMA) and central nervous system demyelination. We present a patient with SMA-like phenotype carrying a homozygous mutation in RBM7-a subunit of the nuclear exosome targeting (NEXT) complex-which is known to bind and carry specific subtypes of coding and non-coding RNAs to the exosome. The NEXT complex with other protein complexes is responsible for the substrate specificity of the exosome. We performed RNA-sequencing (RNA-seq) analysis on primary fibroblasts of patients with mutations in EXOSC8 and RBM7 and gene knock-down experiments using zebrafish as a model system. RNA-seq analysis identified significantly altered expression of 62 transcripts shared by the two patient cell lines. Knock-down of rbm7, exosc8 and exosc3 in zebrafish showed a common pattern of defects in motor neurons and cerebellum. Our data indicate that impaired RNA metabolism may underlie the clinical phenotype by fine tuning gene expression which is essential for correct neuronal differentiation.
Collapse
Affiliation(s)
- Michele Giunta
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
| | - Shimon Edvardson
- The Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center, 91120 Jerusalem, Israel
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
| | - Yaobo Xu
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
| | - Markus Schuelke
- Department of Neuropediatrics and NeuroCure Clinical Research Center, Charité-Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| | - Aurora Gomez-Duran
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
| | - Veronika Boczonadi
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
| | - Orly Elpeleg
- The Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Juliane S Müller
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
| | - Rita Horvath
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
- Institute of Genetic Medicine, Newcastle University, Central Parkway, NE1 3BZ, Newcastle upon Tyne, UK
| |
Collapse
|
32
|
Suto N, Mieda T, Iizuka A, Nakamura K, Hirai H. Morphological and Functional Attenuation of Degeneration of Peripheral Neurons by Mesenchymal Stem Cell-Conditioned Medium in Spinocerebellar Ataxia Type 1-Knock-in Mice. CNS Neurosci Ther 2016; 22:670-6. [PMID: 27140210 DOI: 10.1111/cns.12560] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/13/2022] Open
Abstract
AIMS Spinocerebellar ataxia type 1 (SCA1) is caused by the ataxin-1 protein (ATXN1) with an abnormally expanded polyglutamine tract and is characterized by progressive neurodegeneration. We previously showed that intrathecal injection of mesenchymal stem cells (MSCs) during the nonsymptomatic stage mitigates the degeneration of the peripheral nervous system (PNS) neurons in SCA1-knock-in (SCA1-KI) mice. We tested in this study whether the therapeutic effects of MSCs in SCA1-KI mice could be reproduced with MSC-releasing factor(s). METHODS To test the effects of MSC-releasing factor(s), we used MSC-conditioned medium (MSC-CM). MSC-CM was intrathecally and/or intravenously injected into young SCA1-KI mice, and the therapeutic effects were assessed in the PNS at later ages using immunostaining, electrophysiology, and behavioral tests. RESULTS MSC-CM attenuated the degeneration of axons and myelin of spinal motor neurons. Consequently, the injected SCA1-KI mice exhibited smaller reductions in nerve conduction velocity in spinal motor neurons and reduced motor incoordination than the untreated mice. CONCLUSIONS These results suggest that factors released from MSC mitigate the morphological and functional abnormalities in the PNS that are observed in SCA1-KI mice in a paracrine manner.
Collapse
Affiliation(s)
- Nana Suto
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Tokue Mieda
- Department of Orthopedic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Akira Iizuka
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kazuhiro Nakamura
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
33
|
Mieda T, Suto N, Iizuka A, Matsuura S, Iizuka H, Takagishi K, Nakamura K, Hirai H. Mesenchymal stem cells attenuate peripheral neuronal degeneration in spinocerebellar ataxia type 1 knockin mice. J Neurosci Res 2015; 94:246-52. [PMID: 26707550 DOI: 10.1002/jnr.23698] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 09/02/2015] [Accepted: 11/04/2015] [Indexed: 12/14/2022]
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a devastating neurodegenerative disorder in which an abnormally expanded polyglutamine tract is inserted into causative ataxin-1 proteins. We have previously shown that SCA1 knockin (SCA1-KI) mice over 6 months of age exhibit a degeneration of motor neuron axons and their encasing myelin sheaths, as reported in SCA1 patients. We examined whether axon degeneration precedes myelin degeneration or vice versa in SCA1-KI mice and then attempted to mitigate motor neuron degeneration by intrathecally administering mesenchymal stem cells (MSCs). Temporal examination of the diameters of motor neuron axons and their myelin sheaths revealed a decrease in diameter of the axon but not of the myelin sheaths in SCA1-KI mice as early as 1 month of age, which suggests secondary degeneration of the myelin sheaths. We injected MSCs into the intrathecal space of SCA1-KI mice at 1 month of age, which resulted in a significant suppression of degeneration of both motor neuron axons and myelin sheaths, even 6 months after the MSC injection. Thus, MSCs effectively suppressed peripheral nervous system degeneration in SCA1-KI mice. It has not yet been clarified how clinically administered MSCs exhibit significant therapeutic effects in patients with SCA1. The morphological evidence presented in this current mouse study might explain the mechanisms that underlie the therapeutic effects of MSCs that are observed in patients with SCA1.
Collapse
Affiliation(s)
- Tokue Mieda
- Department of Orthopedic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Nana Suto
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Akira Iizuka
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Serina Matsuura
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Haku Iizuka
- Department of Orthopedic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kenji Takagishi
- Department of Orthopedic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Kazuhiro Nakamura
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
34
|
Nakamura K, Mieda T, Suto N, Matsuura S, Hirai H. Mesenchymal stem cells as a potential therapeutic tool for spinocerebellar ataxia. THE CEREBELLUM 2015; 14:165-70. [PMID: 25280585 DOI: 10.1007/s12311-014-0604-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Spinocerebellar ataxia (SCA) is a devastating progressive neurodegenerative disorder, for which no effective treatments have been developed. However, some studies have shown that an intracerebellar or intrathecal injection of mesenchymal stem cells (MSCs) was partially effective in some genetic mouse models of cerebellar ataxia such as SCA1 and Lurcher mutant. MSCs likely exert their therapeutic efficacy by secreting innate factors to induce neuronal growth and synaptic connection and reduce apoptosis. In this review, we introduce the therapeutic influence of MSCs on each mouse model for cerebellar ataxia and the possible mechanisms underlying the action of MSCs. We also introduce studies on the safety and effectiveness of umbilical cord MSCs for patients with SCA.
Collapse
Affiliation(s)
- Kazuhiro Nakamura
- Department of Neurophysiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | | | | | | | | |
Collapse
|
35
|
Pedroso JL, de Souza PVS, Pinto WBVDR, Braga-Neto P, Albuquerque MVC, Saraiva-Pereira ML, Jardim LB, Barsottini OGP. SCA1 patients may present as hereditary spastic paraplegia and must be included in spastic-ataxias group. Parkinsonism Relat Disord 2015; 21:1243-6. [DOI: 10.1016/j.parkreldis.2015.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/04/2015] [Accepted: 07/19/2015] [Indexed: 11/30/2022]
|
36
|
Hearst SM, Shao Q, Lopez M, Raucher D, Vig PJS. Focused cerebellar laser light induced hyperthermia improves symptoms and pathology of polyglutamine disease SCA1 in a mouse model. THE CEREBELLUM 2015; 13:596-606. [PMID: 24930030 DOI: 10.1007/s12311-014-0576-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Spinocerebellar ataxia 1 (SCA1) results from pathologic glutamine expansion in the ataxin-1 protein (ATXN1). This misfolded ATXN1 causes severe Purkinje cell (PC) loss and cerebellar ataxia in both humans and mice with the SCA1 disease. The molecular chaperone heat-shock proteins (HSPs) are known to modulate polyglutamine protein aggregation and are neuroprotective. Since HSPs are induced under stress, we explored the effects of focused laser light induced hyperthermia (HT) on HSP-mediated protection against ATXN1 toxicity. We first tested the effects of HT in a cell culture model and found that HT induced Hsp70 and increased its localization to nuclear inclusions in HeLa cells expressing GFP-ATXN1[82Q]. HT treatment decreased ATXN1 aggregation by making GFP-ATXN1[82Q] inclusions smaller and more numerous compared to non-treated cells. Further, we tested our HT approach in vivo using a transgenic (Tg) mouse model of SCA1. We found that our laser method increased cerebellar temperature from 38 to 40 °C without causing any neuronal damage or inflammatory response. Interestingly, mild cerebellar HT stimulated the production of Hsp70 to a significant level. Furthermore, multiple exposure of focused cerebellar laser light induced HT to heterozygous SCA1 transgenic (Tg) mice significantly suppressed the SCA1 phenotype as compared to sham-treated control animals. Moreover, in treated SCA1 Tg mice, the levels of PC calcium signaling/buffering protein calbindin-D28k markedly increased followed by a reduction in PC neurodegenerative morphology. Taken together, our data suggest that laser light induced HT is a novel non-invasive approach to treat SCA1 and maybe other polyglutamine disorders.
Collapse
Affiliation(s)
- Scoty M Hearst
- Department of Neurology, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA
| | | | | | | | | |
Collapse
|
37
|
Long-term oral administration of the NMDA receptor antagonist memantine extends life span in spinocerebellar ataxia type 1 knock-in mice. Neurosci Lett 2015; 592:37-41. [PMID: 25725171 DOI: 10.1016/j.neulet.2015.02.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 11/20/2022]
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a progressive neurodegenerative disease caused by extension of a CAG repeat in the Sca1gene. Although the mechanisms underlying the symptoms of SCA1 have not been determined, aberrant neuronal activation potentially contributes to the neuronal cell death characteristic of the disease. Here we examined the potential involvement of extrasynaptic N-methyl-d-aspartate receptor (NMDAR) activation in the pathogenesis of SCA1 by administering memantine, a low-affinity noncompetitive NMDAR antagonist, in SCA1 knock-in (KI) mice. In KI mice, the exon in the ataxin 1 gene is replaced with abnormally expanded 154CAG repeats. Memantine was administered orally to the SCA1 KI mice from 4 weeks of age until death. The treatment significantly attenuated body-weight loss and prolonged the life span of SCA1 KI mice. Furthermore, memantine significantly suppressed the loss of Purkinje cells in the cerebellum and motor neurons in the dorsal motor nucleus of the vagus, which are critical for motor function and parasympathetic function, respectively. These findings support the contribution of aberrant activation of extrasynaptic NMDARs to neuronal cell death in SCA1 KI mice and suggest that memantine may also have therapeutic benefits in human SCA1 patients.
Collapse
|
38
|
Vig PJS, Hearst SM, Shao Q, Lopez ME. Knockdown of acid-sensing ion channel 1a (ASIC1a) suppresses disease phenotype in SCA1 mouse model. THE CEREBELLUM 2015; 13:479-90. [PMID: 24788087 DOI: 10.1007/s12311-014-0563-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The mutated ataxin-1 protein in spinocerebellar ataxia 1 (SCA1) targets Purkinje cells (PCs) of the cerebellum and causes progressive ataxia due to loss of PCs and neurons of the brainstem. The exact mechanism of this cellular loss is still not clear. Currently, there are no treatments for SCA1; however, understanding of the mechanisms that regulate SCA1 pathology is essential for devising new therapies for SCA1 patients. We previously established a connection between the loss of intracellular calcium-buffering and calcium-signalling proteins with initiation of neurodegeneration in SCA1 transgenic (Tg) mice. Recently, acid-sensing ion channel 1a (ASIC1a) have been implicated in calcium-mediated toxicity in many brain disorders. Here, we report generating SCA1 Tg mice in the ASIC1a knockout (KO) background and demonstrate that the deletion of ASIC1a gene expression causes suppression of the SCA1 disease phenotype. Loss of the ASIC1a channel in SCA1/ASIC1a KO mice resulted in the improvement of motor deficit and decreased PC degeneration. Interestingly, the expression of the ASIC1 variant, ASIC1b, was upregulated in the cerebellum of both SCA1/ASIC1a KO and ASIC1a KO animals as compared to the wild-type (WT) and SCA1 Tg mice. Further, these SCA1/ASIC1a KO mice exhibited translocation of PC calcium-binding protein calbindin-D28k from the nucleus to the cytosol in young animals, which otherwise have both cytosolic and nuclear localization. Furthermore, in addition to higher expression of calcium-buffering protein parvalbumin, PCs of the older SCA1/ASIC1a KO mice showed a decrease in morphologic abnormalities as compared to the age-matched SCA1 animals. Our data suggest that ASIC1a may be a mediator of SCA1 pathogenesis and targeting ASIC1a could be a novel approach to treat SCA1.
Collapse
Affiliation(s)
- Parminder J S Vig
- Department of Neurology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA,
| | | | | | | |
Collapse
|
39
|
Fan HC, Ho LI, Chi CS, Chen SJ, Peng GS, Chan TM, Lin SZ, Harn HJ. Polyglutamine (PolyQ) diseases: genetics to treatments. Cell Transplant 2015; 23:441-58. [PMID: 24816443 DOI: 10.3727/096368914x678454] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The polyglutamine (polyQ) diseases are a group of neurodegenerative disorders caused by expanded cytosine-adenine-guanine (CAG) repeats encoding a long polyQ tract in the respective proteins. To date, a total of nine polyQ disorders have been described: six spinocerebellar ataxias (SCA) types 1, 2, 6, 7, 17; Machado-Joseph disease (MJD/SCA3); Huntington's disease (HD); dentatorubral pallidoluysian atrophy (DRPLA); and spinal and bulbar muscular atrophy, X-linked 1 (SMAX1/SBMA). PolyQ diseases are characterized by the pathological expansion of CAG trinucleotide repeat in the translated region of unrelated genes. The translated polyQ is aggregated in the degenerated neurons leading to the dysfunction and degeneration of specific neuronal subpopulations. Although animal models of polyQ disease for understanding human pathology and accessing disease-modifying therapies in neurodegenerative diseases are available, there is neither a cure nor prevention for these diseases, and only symptomatic treatments for polyQ diseases currently exist. Long-term pharmacological treatment is so far disappointing, probably due to unwanted complications and decreasing drug efficacy. Cellular transplantation of stem cells may provide promising therapeutic avenues for restoration of the functions of degenerative and/or damaged neurons in polyQ diseases.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Mesenchymal stem cells ameliorate cerebellar pathology in a mouse model of spinocerebellar ataxia type 1. THE CEREBELLUM 2014; 13:323-30. [PMID: 24242763 DOI: 10.1007/s12311-013-0536-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a progressive neurodegenerative disorder caused by the expansion of a polyglutamine tract in the ataxin-1 protein. To date, no fundamental treatments for SCA1 have been elucidated. However, some studies have shown that mesenchymal stem cells (MSCs) are partially effective in other genetic mouse models of cerebellar ataxia. In this study, we tested the efficacy of the intrathecal injection of MSCs in the treatment of SCA1 in transgenic (SCA1-Tg) mice. We found that intrathecal injection of only 3 × 10(3) MSCs greatly mitigated the cerebellar neuronal disorganization observed in SCA1 transgenic mice (SCA1-Tg mice). Although the Purkinje cells (PCs) of 24-week-old nontreated SCA1-Tg mice displayed a multilayer arrangement, SCA1-Tg mice at a similar age injected with MSCs displayed monolayer PCs. Furthermore, intrathecal injection of MSCs suppressed the atrophy of PC dendrites in SCA1-Tg mice. Finally, behavioral tests demonstrated that MSCs normalized deficits in motor coordination in SCA1-Tg mice. Future studies should be performed to develop optimal protocols for intrathecal transplantation of MSCs in SCA1 model primates with the aim of developing applications for SCA1 patients.
Collapse
|
41
|
Abstract
Heredoataxias are a group of genetic disorders with a cerebellar syndrome as the leading clinical manifestation. The current classification distinguishes heredoataxias according to the trait of inheritance into autosomal dominant, autosomal recessive, X-linked, and maternally inherited heredoataxias. The autosomal dominant heredoataxias are separated into spinocerebellar ataxias (SCA1-8, 10-15, 17-23, 25-30, and dentato-rubro-pallido-luysian atrophy), episodic ataxias (EA1-7), and autosomal dominant mitochondrial heredoataxias (Leigh syndrome, MIRAS, ADOAD, and AD-CPEO). The autosomal recessive ataxias are separated into Friedreich ataxia, ataxia due to vitamin E deficiency, ataxia due to Abeta-lipoproteinemia, Refsum disease, late-onset Tay-Sachs disease, cerebrotendineous xanthomatosis, spinocerebellar ataxia with axonal neuropathy, ataxia telangiectasia, ataxia telangiectasia-like disorder, ataxia with oculomotor apraxia 1 and 2, spastic ataxia of Charlevoix-Saguenay, Cayman ataxia, Marinesco-Sjögren syndrome, and autosomal recessive mitochondrial ataxias (AR-CPEO, SANDO, SCAE, AHS, IOSCA, MEMSA, LBSL CoQ-deficiency, PDC-deficiency). Only two of the heredoataxias, fragile X/tremor/ataxia syndrome, and XLSA/A are transmitted via an X-linked trait. Maternally inherited heredoataxias are due to point mutations in genes encoding for tRNAs, rRNAs, respiratory chain subunits or single large scale deletions/duplications of the mitochondrial DNA and include MELAS, MERRF, KSS, PS, MILS, NARP, and non-syndromic mitochondrial disorders. Treatment of heredoataxias is symptomatic and supportive and may have a beneficial effect in single patients.**Please see page 424 for abbreviation list.
Collapse
|
42
|
Penetrating the cell membrane, thermal targeting and novel anticancer drugs: the development of thermally targeted, elastin-like polypeptide cancer therapeutics. Ther Deliv 2014; 5:429-45. [PMID: 24856169 DOI: 10.4155/tde.14.14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Therapeutic peptides offer important cancer treatment approaches. Designed to inhibit oncogenes and other oncoproteins, early therapeutic peptides applications were hampered by pharmacokinetic properties now addressed through tumor targeting strategies. Active targeting with environmentally responsive biopolymers or macromolecules enhances therapeutics accumulation at tumor sites; passive targeting with macromolecules, or liposomes, exploits angiogenesis and poor lymphatic drainage to preferentially accumulate therapeutics within tumors. Genetically engineered, thermally-responsive, elastin-like polypeptides use both strategies and cell-penetrating peptides to further intratumoral cell uptake. This review describes the development and application of cell-penetrating peptide-elastin-like polypeptide therapeutics for the thermally targeted delivery of therapeutic peptides.
Collapse
|
43
|
Metabolic evidence for cerebral neurodegeneration in spinocerebellar ataxia type 1. THE CEREBELLUM 2014; 13:199-206. [PMID: 24085647 DOI: 10.1007/s12311-013-0527-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Autosomal-dominant spinocerebellar ataxia type 1 (SCA1) is an adult-onset progressive disorder with well-characterized neurodegeneration in the cerebellum and brainstem. The objective of this study is to evaluate neurochemical changes associated with neurodegeneration in cerebral tissue in SCA1 patients compared to age- and gender-matched healthy controls. Nine patients with genetically proven SCA1 and nine gender- and age-matched healthy controls were prospectively recruited from the ataxia clinic and received clinical examination. A 1.5 T single-voxel brain proton MR spectroscopy was performed for total N-acetyl aspartate (tNAA) in cerebellum, parietofrontal lobe white matter, sensory cortex, and visual cortex. In the patients, tNAA was severely decreased in the cerebellar voxel; however, in the voxels positioned in sensory cortex, parietofrontal lobe white matter and visual cortex tNAA was reduced in comparison to controls. In addition to the profoundly affected cerebellum, we also found evidence for cerebral neurodegeneration in parietal lobe white matter, sensory cortex, and visual cortex in SCA1 patients illustrating a multisystem neurodegenerative character of the disease.
Collapse
|
44
|
Koscianska E, Krzyzosiak WJ. Current understanding of the role of microRNAs in spinocerebellar ataxias. CEREBELLUM & ATAXIAS 2014; 1:7. [PMID: 26331031 PMCID: PMC4552431 DOI: 10.1186/2053-8871-1-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/30/2014] [Indexed: 12/21/2022]
Abstract
The number of studies highlighting the role of microRNAs (miRNAs) in human physiology and diseases is growing, but many miRNA-driven regulatory mechanisms remain elusive. A proper understanding of the exact functions of individual miRNAs and their interaction with specific targets is vitally important because such knowledge might help cure diseases for which no effective treatment currently exists. Herein, we present current views on the role of the miRNA-mediated regulation of gene expression in the case of select spinocerebellar ataxias (SCAs) and their potential involvement in the pathogenesis of these diseases. Specifically, we summarize published data showing the known links between miRNAs and CAG repeat-dependent SCAs. Moreover, using the example of SCA type 3 (SCA3), we refer to the issue of prediction and validation of miRNA targets, and we demonstrate that miR-181a-1 may regulate the 3'-UTR of the ATXN3 gene.
Collapse
Affiliation(s)
- Edyta Koscianska
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str, 61-704 Poznan, Poland
| | - Wlodzimierz J Krzyzosiak
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str, 61-704 Poznan, Poland
| |
Collapse
|
45
|
Hearst SM, Shao Q, Lopez M, Raucher D, Vig PJS. The design and delivery of a PKA inhibitory polypeptide to treat SCA1. J Neurochem 2014; 131:101-14. [PMID: 24903464 DOI: 10.1111/jnc.12782] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 05/13/2014] [Accepted: 06/02/2014] [Indexed: 01/28/2023]
Abstract
Spinocerebellar ataxia-1 (SCA1) is a neurodegenerative disease that primarily targets Purkinje cells (PCs) of the cerebellum. The exact mechanism of PC degeneration is unknown, however, it is widely believed that mutant ataxin-1 becomes toxic because of the phosphorylation of its serine 776 (S776) residue by cAMP-dependent protein kinase A (PKA). Therefore, to directly modulate mutant ATXN1 S776 phosphorylation and aggregation, we designed a therapeutic polypeptide to inhibit PKA. This polypeptide comprised of a thermally responsive elastin-like peptide (ELP) carrier, which increases peptide half-life, a PKA inhibitory peptide (PKI), and a cell-penetrating peptide (Synb1). We observed that our therapeutic polypeptide, Synb1-ELP-PKI, inhibited PKA activity at concentrations similar to the PKI peptide. Additionally, Synb1-ELP-PKI significantly suppressed mutant ATXN1 S776 phosphorylation and intranuclear inclusion formation in cell culture. Further, Synb1-ELP-PKI treatment improved SCA1 PC morphology in cerebellar slice cultures. Furthermore, the Synb1-ELP peptide carrier crossed the blood-brain barrier and localized to the cerebellum via the i.p. or intranasal route. Here, we show the intranasal delivery of ELP-based peptides to the brain as a novel delivery strategy. We also demonstrate that our therapeutic polypeptide has a great potential to target the neurotoxic S776 phosphorylation pathway in the SCA1 disease.
Collapse
Affiliation(s)
- Scoty M Hearst
- Department of Neurology, University of Mississippi Medical Center, Jackson, Mississippi, USA; Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | | | | | | |
Collapse
|
46
|
Matilla-Dueñas A, Ashizawa T, Brice A, Magri S, McFarland KN, Pandolfo M, Pulst SM, Riess O, Rubinsztein DC, Schmidt J, Schmidt T, Scoles DR, Stevanin G, Taroni F, Underwood BR, Sánchez I. Consensus paper: pathological mechanisms underlying neurodegeneration in spinocerebellar ataxias. CEREBELLUM (LONDON, ENGLAND) 2014; 13:269-302. [PMID: 24307138 PMCID: PMC3943639 DOI: 10.1007/s12311-013-0539-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intensive scientific research devoted in the recent years to understand the molecular mechanisms or neurodegeneration in spinocerebellar ataxias (SCAs) are identifying new pathways and targets providing new insights and a better understanding of the molecular pathogenesis in these diseases. In this consensus manuscript, the authors discuss their current views on the identified molecular processes causing or modulating the neurodegenerative phenotype in spinocerebellar ataxias with the common opinion of translating the new knowledge acquired into candidate targets for therapy. The following topics are discussed: transcription dysregulation, protein aggregation, autophagy, ion channels, the role of mitochondria, RNA toxicity, modulators of neurodegeneration and current therapeutic approaches. Overall point of consensus includes the common vision of neurodegeneration in SCAs as a multifactorial, progressive and reversible process, at least in early stages. Specific points of consensus include the role of the dysregulation of protein folding, transcription, bioenergetics, calcium handling and eventual cell death with apoptotic features of neurons during SCA disease progression. Unresolved questions include how the dysregulation of these pathways triggers the onset of symptoms and mediates disease progression since this understanding may allow effective treatments of SCAs within the window of reversibility to prevent early neuronal damage. Common opinions also include the need for clinical detection of early neuronal dysfunction, for more basic research to decipher the early neurodegenerative process in SCAs in order to give rise to new concepts for treatment strategies and for the translation of the results to preclinical studies and, thereafter, in clinical practice.
Collapse
Affiliation(s)
- A Matilla-Dueñas
- Health Sciences Research Institute Germans Trias i Pujol (IGTP), Ctra. de Can Ruti, Camí de les Escoles s/n, Badalona, Barcelona, Spain,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Menon RP, Soong D, de Chiara C, Holt M, McCormick JE, Anilkumar N, Pastore A. Mapping the self-association domains of ataxin-1: identification of novel non overlapping motifs. PeerJ 2014; 2:e323. [PMID: 24711972 PMCID: PMC3970802 DOI: 10.7717/peerj.323] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/07/2014] [Indexed: 12/13/2022] Open
Abstract
The neurodegenerative disease spinocerebellar ataxia type 1 (SCA1) is caused by aggregation and misfolding of the ataxin-1 protein. While the pathology correlates with mutations that lead to expansion of a polyglutamine tract in the protein, other regions contribute to the aggregation process as also non-expanded ataxin-1 is intrinsically aggregation-prone and forms nuclear foci in cell. Here, we have used a combined approach based on FRET analysis, confocal microscopy and in vitro techniques to map aggregation-prone regions other than polyglutamine and to establish the importance of dimerization in self-association/foci formation. Identification of aggregation-prone regions other than polyglutamine could greatly help the development of SCA1 treatment more specific than that based on targeting the low complexity polyglutamine region.
Collapse
Affiliation(s)
- Rajesh P Menon
- MRC National Institute for Medical Research, The Ridgeway , London , UK
| | - Daniel Soong
- Randall Division for Cell and Molecular Biophysics, New Hunt's House, King's College London , Guy's Campus, London , UK ; British Heart Foundation Centre of Research Excellence, King's College London , Denmark Hill Campus, London , UK
| | - Cesira de Chiara
- MRC National Institute for Medical Research, The Ridgeway , London , UK
| | - Mark Holt
- Randall Division for Cell and Molecular Biophysics, New Hunt's House, King's College London , Guy's Campus, London , UK
| | - John E McCormick
- MRC National Institute for Medical Research, The Ridgeway , London , UK
| | - Narayana Anilkumar
- British Heart Foundation Centre of Research Excellence, King's College London , Denmark Hill Campus, London , UK
| | - Annalisa Pastore
- MRC National Institute for Medical Research, The Ridgeway , London , UK ; Department of Molecular Neuroscience, Institute of Psychiatry, King's College London , Denmark Hill Campus, London , UK
| |
Collapse
|
48
|
Increased Catabolic State in Spinocerebellar Ataxia Type 1 Patients. THE CEREBELLUM 2014; 13:440-6. [DOI: 10.1007/s12311-014-0555-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Functionally enigmatic genes: a case study of the brain ignorome. PLoS One 2014; 9:e88889. [PMID: 24523945 PMCID: PMC3921226 DOI: 10.1371/journal.pone.0088889] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 01/14/2014] [Indexed: 12/26/2022] Open
Abstract
What proportion of genes with intense and selective expression in specific tissues, cells, or systems are still almost completely uncharacterized with respect to biological function? In what ways do these functionally enigmatic genes differ from well-studied genes? To address these two questions, we devised a computational approach that defines so-called ignoromes. As proof of principle, we extracted and analyzed a large subset of genes with intense and selective expression in brain. We find that publications associated with this set are highly skewed--the top 5% of genes absorb 70% of the relevant literature. In contrast, approximately 20% of genes have essentially no neuroscience literature. Analysis of the ignorome over the past decade demonstrates that it is stubbornly persistent, and the rapid expansion of the neuroscience literature has not had the expected effect on numbers of these genes. Surprisingly, ignorome genes do not differ from well-studied genes in terms of connectivity in coexpression networks. Nor do they differ with respect to numbers of orthologs, paralogs, or protein domains. The major distinguishing characteristic between these sets of genes is date of discovery, early discovery being associated with greater research momentum--a genomic bandwagon effect. Finally we ask to what extent massive genomic, imaging, and phenotype data sets can be used to provide high-throughput functional annotation for an entire ignorome. In a majority of cases we have been able to extract and add significant information for these neglected genes. In several cases--ELMOD1, TMEM88B, and DZANK1--we have exploited sequence polymorphisms, large phenome data sets, and reverse genetic methods to evaluate the function of ignorome genes.
Collapse
|
50
|
Pedroso JL, Barsottini OGP. Spinal cord atrophy in spinocerebellar ataxia type 1. ARQUIVOS DE NEURO-PSIQUIATRIA 2013; 71:977. [DOI: 10.1590/0004-282x20130187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 06/25/2013] [Indexed: 11/22/2022]
|