1
|
Jackson NN, Stagray JA, Snell HD. Cerebellar contributions to dystonia: unraveling the role of Purkinje cells and cerebellar nuclei. DYSTONIA (LAUSANNE, SWITZERLAND) 2025; 4:14006. [PMID: 40115904 PMCID: PMC11925549 DOI: 10.3389/dyst.2025.14006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Dystonias are a group of neurodegenerative disorders that result in altered physiology associated with motor movements. Both the basal ganglia and the cerebellum, brain regions involved in motor learning, sensory perception integration, and reward, have been implicated in the pathology of dystonia, but the cellular and subcellular mechanisms remain diverse and for some forms of dystonia, elusive. The goal of the current review is to summarize recent evidence of cerebellar involvement in different subtypes of dystonia with a focus on Purkinje cell (PC) and cerebellar nuclei (CN) dysfunction, to find commonalities in the pathology that could lay the groundwork for the future development of therapeutics for patients with dystonia. Here we will briefly discuss the physical and functional connections between the basal ganglia and the cerebellum and how these connections could contribute to dystonic symptoms. We proceed to use human and animal model data to discuss the contributions of cerebellar cell types to specific dystonias and movement disorders where dystonia is a secondary symptom. Ultimately, we suggest PC and CN irregularity could be a locus for dystonia through impaired calcium dynamics.
Collapse
Affiliation(s)
- Nichelle N Jackson
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| | - Jacob A Stagray
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| | - Heather D Snell
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
- Wu Tsai Institute, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
2
|
Lee JH. Oxidative stress and the multifaceted roles of ATM in maintaining cellular redox homeostasis. Redox Biol 2024; 75:103269. [PMID: 39018798 PMCID: PMC11301354 DOI: 10.1016/j.redox.2024.103269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024] Open
Abstract
The ataxia telangiectasia mutated (ATM) protein kinase is best known as a master regulator of the DNA damage response. However, accumulating evidence has unveiled an equally vital function for ATM in sensing oxidative stress and orchestrating cellular antioxidant defenses to maintain redox homeostasis. ATM can be activated through a non-canonical pathway involving intermolecular disulfide crosslinking of the kinase dimers, distinct from its canonical activation by DNA double-strand breaks. Structural studies have elucidated the conformational changes that allow ATM to switch into an active redox-sensing state upon oxidation. Notably, loss of ATM function results in elevated reactive oxygen species (ROS) levels, altered antioxidant profiles, and mitochondrial dysfunction across multiple cell types and tissues. This oxidative stress arising from ATM deficiency has been implicated as a central driver of the neurodegenerative phenotypes in ataxia-telangiectasia (A-T) patients, potentially through mechanisms involving oxidative DNA damage, PARP hyperactivation, and widespread protein aggregation. Moreover, defective ATM oxidation sensing disrupts transcriptional programs and RNA metabolism, with detrimental impacts on neuronal homeostasis. Significantly, antioxidant therapy can ameliorate cellular and organismal abnormalities in various ATM-deficient models. This review synthesizes recent advances illuminating the multifaceted roles of ATM in preserving redox balance and mitigating oxidative insults, providing a unifying paradigm for understanding the complex pathogenesis of A-T disease.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
3
|
Sekerková G, Kilic S, Cheng YH, Fredrick N, Osmani A, Kim H, Opal P, Martina M. Phenotypical, genotypical and pathological characterization of the moonwalker mouse, a model of ataxia. Neurobiol Dis 2024; 195:106492. [PMID: 38575093 PMCID: PMC11089908 DOI: 10.1016/j.nbd.2024.106492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/13/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024] Open
Abstract
We performed a comprehensive study of the morphological, functional, and genetic features of moonwalker (MWK) mice, a mouse model of spinocerebellar ataxia caused by a gain of function of the TRPC3 channel. These mice show numerous behavioral symptoms including tremor, altered gait, circling behavior, impaired motor coordination, impaired motor learning and decreased limb strength. Cerebellar pathology is characterized by early and almost complete loss of unipolar brush cells as well as slowly progressive, moderate loss of Purkinje cell (PCs). Structural damage also includes loss of synaptic contacts from parallel fibers, swollen ER structures, and degenerating axons. Interestingly, no obvious correlation was observed between PC loss and severity of the symptoms, as the phenotype stabilizes around 2 months of age, while the cerebellar pathology is progressive. This is probably due to the fact that PC function is severely impaired much earlier than the appearance of PC loss. Indeed, PC firing is already impaired in 3 weeks old mice. An interesting feature of the MWK pathology that still remains to be explained consists in a strong lobule selectivity of the PC loss, which is puzzling considering that TRPC is expressed in every PC. Intriguingly, genetic analysis of MWK cerebella shows, among other alterations, changes in the expression of both apoptosis inducing and resistance factors possibly suggesting that damaged PCs initiate specific cellular pathways that protect them from overt cell loss.
Collapse
Affiliation(s)
- Gabriella Sekerková
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA.
| | - Sumeyra Kilic
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Yen-Hsin Cheng
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Natalie Fredrick
- Department of Neurology, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Anne Osmani
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Haram Kim
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Puneet Opal
- Department of Neurology, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Marco Martina
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA.
| |
Collapse
|
4
|
Paull TT, Woolley PR. A-T neurodegeneration and DNA damage-induced transcriptional stress. DNA Repair (Amst) 2024; 135:103647. [PMID: 38377644 PMCID: PMC11707827 DOI: 10.1016/j.dnarep.2024.103647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/08/2024] [Indexed: 02/22/2024]
Abstract
Loss of the ATM protein kinase in humans results in Ataxia-telangiectasia, a disorder characterized by childhood-onset neurodegeneration of the cerebellum as well as cancer predisposition and immunodeficiency. Although many aspects of ATM function are well-understood, the mechanistic basis of the progressive cerebellar ataxia that occurs in patients is not. Here we review recent progress related to the role of ATM in neurons and the cerebellum that comes from many sources: animal models, post-mortem brain tissue samples, and human neurons in culture. These observations have revealed new insights into the consequences of ATM loss on DNA damage, gene expression, and immune signaling in the brain. Many results point to the importance of reactive oxygen species as well as single-strand DNA breaks in the progression of molecular events leading to neuronal dysfunction. In addition, innate immunity signaling pathways appear to play a critical role in ATM functions in microglia, responding to various forms of nucleic acid sensors and regulating survival of neurons and other cell types. Overall, the results lead to an updated view of transcriptional stress and DNA damage resulting from ATM loss that results in changes in gene expression as well as neuroinflammation that contribute to the cerebellar neurodegeneration observed in patients.
Collapse
Affiliation(s)
- Tanya T Paull
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA.
| | - Phillip R Woolley
- The University of Texas at Austin, Department of Molecular Biosciences, Austin, TX 78712, USA
| |
Collapse
|
5
|
Izquierdo-Villalba I, Mirra S, Manso Y, Parcerisas A, Rubio J, Del Valle J, Gil-Bea FJ, Ulloa F, Herrero-Lorenzo M, Verdaguer E, Benincá C, Castro-Torres RD, Rebollo E, Marfany G, Auladell C, Navarro X, Enríquez JA, López de Munain A, Soriano E, Aragay AM. A mammalian-specific Alex3/Gα q protein complex regulates mitochondrial trafficking, dendritic complexity, and neuronal survival. Sci Signal 2024; 17:eabq1007. [PMID: 38320000 DOI: 10.1126/scisignal.abq1007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 01/16/2024] [Indexed: 02/08/2024]
Abstract
Mitochondrial dynamics and trafficking are essential to provide the energy required for neurotransmission and neural activity. We investigated how G protein-coupled receptors (GPCRs) and G proteins control mitochondrial dynamics and trafficking. The activation of Gαq inhibited mitochondrial trafficking in neurons through a mechanism that was independent of the canonical downstream PLCβ pathway. Mitoproteome analysis revealed that Gαq interacted with the Eutherian-specific mitochondrial protein armadillo repeat-containing X-linked protein 3 (Alex3) and the Miro1/Trak2 complex, which acts as an adaptor for motor proteins involved in mitochondrial trafficking along dendrites and axons. By generating a CNS-specific Alex3 knockout mouse line, we demonstrated that Alex3 was required for the effects of Gαq on mitochondrial trafficking and dendritic growth in neurons. Alex3-deficient mice had altered amounts of ER stress response proteins, increased neuronal death, motor neuron loss, and severe motor deficits. These data revealed a mammalian-specific Alex3/Gαq mitochondrial complex, which enables control of mitochondrial trafficking and neuronal death by GPCRs.
Collapse
Affiliation(s)
| | - Serena Mirra
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBER-CIBERER), ISCIII, Madrid 28031, Spain
- Institut de Biomedicina- Institut de Recerca Sant Joan de Déu (IBUB-IRSJD), Universitat de Barcelona, Barcelona 08028, Spain
| | - Yasmina Manso
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Antoni Parcerisas
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic, Central University of Catalonia (UVic-UCC); and Tissue Repair and Regeneration Laboratory (TR2Lab), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), 08500 Vic, Spain
| | - Javier Rubio
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Jaume Del Valle
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, Universitat Autonoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Francisco J Gil-Bea
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián 20014, Spain
| | - Fausto Ulloa
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Marina Herrero-Lorenzo
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
| | - Ester Verdaguer
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Cristiane Benincá
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Rubén D Castro-Torres
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
| | - Elena Rebollo
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Gemma Marfany
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBER-CIBERER), ISCIII, Madrid 28031, Spain
- Institut de Biomedicina- Institut de Recerca Sant Joan de Déu (IBUB-IRSJD), Universitat de Barcelona, Barcelona 08028, Spain
| | - Carme Auladell
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Xavier Navarro
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, Universitat Autonoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - José A Enríquez
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
- Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBER-CIBERFES), Madrid 28031, Spain
| | - Adolfo López de Munain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián 20014, Spain
- Neurology Department, Donostia University Hospital, San Sebastián 20014, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Anna M Aragay
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| |
Collapse
|
6
|
Manolaras I, Del Bondio A, Griso O, Reutenauer L, Eisenmann A, Habermann BH, Puccio H. Mitochondrial dysfunction and calcium dysregulation in COQ8A-ataxia Purkinje neurons are rescued by CoQ10 treatment. Brain 2023; 146:3836-3850. [PMID: 36960552 DOI: 10.1093/brain/awad099] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 01/11/2023] [Accepted: 03/06/2023] [Indexed: 03/25/2023] Open
Abstract
COQ8A-ataxia is a rare form of neurodegenerative disorder due to mutations in the COQ8A gene. The encoded mitochondrial protein is involved in the regulation of coenzyme Q10 biosynthesis. Previous studies on the constitutive Coq8a-/- mice indicated specific alterations of cerebellar Purkinje neurons involving altered electrophysiological function and dark cell degeneration. In the present manuscript, we extend our understanding of the contribution of Purkinje neuron dysfunction to the pathology. By generating a Purkinje-specific conditional COQ8A knockout, we demonstrate that loss of COQ8A in Purkinje neurons is the main cause of cerebellar ataxia. Furthermore, through in vivo and in vitro approaches, we show that COQ8A-depleted Purkinje neurons have abnormal dendritic arborizations, altered mitochondria function and intracellular calcium dysregulation. Furthermore, we demonstrate that oxidative phosphorylation, in particular Complex IV, is primarily altered at presymptomatic stages of the disease. Finally, the morphology of primary Purkinje neurons as well as the mitochondrial dysfunction and calcium dysregulation could be rescued by CoQ10 treatment, suggesting that CoQ10 could be a beneficial treatment for COQ8A-ataxia.
Collapse
Affiliation(s)
- Ioannis Manolaras
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of translational medecine and neurogenetics, 67404 Illkirch, France
- Inserm, U1258, 67404 Illkirch, France
- CNRS, UMR7104, 67404 Illkirch, France
- Université de Strasbourg, 67000 Strasbourg, France
| | - Andrea Del Bondio
- Institut Neuromyogène, Pathophysiology and genetics of the neuron and muscle, Inserm U1315, 69008 Lyon, France
- CNRS, Université Claude Bernard Lyon I, UMR 5261, 69008 Lyon, France
| | - Olivier Griso
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of translational medecine and neurogenetics, 67404 Illkirch, France
- Inserm, U1258, 67404 Illkirch, France
- CNRS, UMR7104, 67404 Illkirch, France
- Université de Strasbourg, 67000 Strasbourg, France
| | - Laurence Reutenauer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of translational medecine and neurogenetics, 67404 Illkirch, France
- Inserm, U1258, 67404 Illkirch, France
- CNRS, UMR7104, 67404 Illkirch, France
- Université de Strasbourg, 67000 Strasbourg, France
- Institut Neuromyogène, Pathophysiology and genetics of the neuron and muscle, Inserm U1315, 69008 Lyon, France
- CNRS, Université Claude Bernard Lyon I, UMR 5261, 69008 Lyon, France
| | - Aurélie Eisenmann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of translational medecine and neurogenetics, 67404 Illkirch, France
- Inserm, U1258, 67404 Illkirch, France
- CNRS, UMR7104, 67404 Illkirch, France
- Université de Strasbourg, 67000 Strasbourg, France
| | - Bianca H Habermann
- CNRS, Institut de Biologie du Développement de Marseille (IBDM), UMR7288, Aix-Marseille University, 13009 Marseille, France
| | - Hélène Puccio
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of translational medecine and neurogenetics, 67404 Illkirch, France
- Inserm, U1258, 67404 Illkirch, France
- CNRS, UMR7104, 67404 Illkirch, France
- Université de Strasbourg, 67000 Strasbourg, France
- Institut Neuromyogène, Pathophysiology and genetics of the neuron and muscle, Inserm U1315, 69008 Lyon, France
- CNRS, Université Claude Bernard Lyon I, UMR 5261, 69008 Lyon, France
| |
Collapse
|
7
|
Buijsen RAM, Hu M, Sáez-González M, Notopoulou S, Mina E, Koning W, Gardiner SL, van der Graaf LM, Daoutsali E, Pepers BA, Mei H, van Dis V, Frimat JP, van den Maagdenberg AMJM, Petrakis S, van Roon-Mom WMC. Spinocerebellar Ataxia Type 1 Characteristics in Patient-Derived Fibroblast and iPSC-Derived Neuronal Cultures. Mov Disord 2023; 38:1428-1442. [PMID: 37278528 DOI: 10.1002/mds.29446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/23/2023] [Accepted: 04/20/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by a polyglutamine expansion in the ataxin-1 protein resulting in neuropathology including mutant ataxin-1 protein aggregation, aberrant neurodevelopment, and mitochondrial dysfunction. OBJECTIVES Identify SCA1-relevant phenotypes in patient-specific fibroblasts and SCA1 induced pluripotent stem cells (iPSCs) neuronal cultures. METHODS SCA1 iPSCs were generated and differentiated into neuronal cultures. Protein aggregation and neuronal morphology were evaluated using fluorescent microscopy. Mitochondrial respiration was measured using the Seahorse Analyzer. The multi-electrode array (MEA) was used to identify network activity. Finally, gene expression changes were studied using RNA-seq to identify disease-specific mechanisms. RESULTS Bioenergetics deficits in patient-derived fibroblasts and SCA1 neuronal cultures showed altered oxygen consumption rate, suggesting involvement of mitochondrial dysfunction in SCA1. In SCA1 hiPSC-derived neuronal cells, nuclear and cytoplasmic aggregates were identified similar in localization as aggregates in SCA1 postmortem brain tissue. SCA1 hiPSC-derived neuronal cells showed reduced dendrite length and number of branching points while MEA recordings identified delayed development in network activity in SCA1 hiPSC-derived neuronal cells. Transcriptome analysis identified 1050 differentially expressed genes in SCA1 hiPSC-derived neuronal cells associated with synapse organization and neuron projection guidance, where a subgroup of 151 genes was highly associated with SCA1 phenotypes and linked to SCA1 relevant signaling pathways. CONCLUSIONS Patient-derived cells recapitulate key pathological features of SCA1 pathogenesis providing a valuable tool for the identification of novel disease-specific processes. This model can be used for high throughput screenings to identify compounds, which may prevent or rescue neurodegeneration in this devastating disease. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ronald A M Buijsen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Michel Hu
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Maria Sáez-González
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sofia Notopoulou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Eleni Mina
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Winette Koning
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Sarah L Gardiner
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Linda M van der Graaf
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Elena Daoutsali
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Barry A Pepers
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Hailiang Mei
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Vera van Dis
- Department of Pathology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Pathology, Erasmus Medical Center, Rotterdam, Zuid-Holland, The Netherlands
| | - Jean-Philippe Frimat
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| | - Spyros Petrakis
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Willeke M C van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, Leiden, Zuid-Holland, The Netherlands
| |
Collapse
|
8
|
Fernández de la Torre M, Fiuza-Luces C, Laine-Menéndez S, Delmiro A, Arenas J, Martín MÁ, Lucia A, Morán M. Pathophysiology of Cerebellar Degeneration in Mitochondrial Disorders: Insights from the Harlequin Mouse. Int J Mol Sci 2023; 24:10973. [PMID: 37446148 DOI: 10.3390/ijms241310973] [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: 05/21/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
By means of a proteomic approach, we assessed the pathways involved in cerebellar neurodegeneration in a mouse model (Harlequin, Hq) of mitochondrial disorder. A differential proteomic profile study (iTRAQ) was performed in cerebellum homogenates of male Hq and wild-type (WT) mice 8 weeks after the onset of clear symptoms of ataxia in the Hq mice (aged 5.2 ± 0.2 and 5.3 ± 0.1 months for WT and Hq, respectively), followed by a biochemical validation of the most relevant changes. Additional groups of 2-, 3- and 6-month-old WT and Hq mice were analyzed to assess the disease progression on the proteins altered in the proteomic study. The proteomic analysis showed that beyond the expected deregulation of oxidative phosphorylation, the cerebellum of Hq mice showed a marked astroglial activation together with alterations in Ca2+ homeostasis and neurotransmission, with an up- and downregulation of GABAergic and glutamatergic neurotransmission, respectively, and the downregulation of cerebellar "long-term depression", a synaptic plasticity phenomenon that is a major player in the error-driven learning that occurs in the cerebellar cortex. Our study provides novel insights into the mechanisms associated with cerebellar degeneration in the Hq mouse model, including a complex deregulation of neuroinflammation, oxidative phosphorylation and glutamate, GABA and amino acids' metabolism.
Collapse
Affiliation(s)
- Miguel Fernández de la Torre
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
| | - Carmen Fiuza-Luces
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
| | - Sara Laine-Menéndez
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
| | - Aitor Delmiro
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
- Servicio de Bioquímica Clínica, Hospital Universitario "12 de Octubre", 28041 Madrid, Spain
| | - Joaquín Arenas
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
| | - Miguel Ángel Martín
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
- Servicio de Genética, Hospital Universitario "12 de Octubre", 28041 Madrid, Spain
| | - Alejandro Lucia
- Faculty of Sports Sciences, European University of Madrid, 28670 Madrid, Spain
- Spanish Network for Biomedical Research in Fragility and Healthy Aging (CIBERFES), 28029 Madrid, Spain
| | - María Morán
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
| |
Collapse
|
9
|
Kapfhammer JP, Shimobayashi E. Viewpoint: spinocerebellar ataxias as diseases of Purkinje cell dysfunction rather than Purkinje cell loss. Front Mol Neurosci 2023; 16:1182431. [PMID: 37426070 PMCID: PMC10323145 DOI: 10.3389/fnmol.2023.1182431] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/22/2023] [Indexed: 07/11/2023] Open
Abstract
Spinocerebellar ataxias (SCAs) are a group of hereditary neurodegenerative diseases mostly affecting cerebellar Purkinje cells caused by a wide variety of different mutations. One subtype, SCA14, is caused by mutations of Protein Kinase C gamma (PKCγ), the dominant PKC isoform present in Purkinje cells. Mutations in the pathway in which PKCγ is active, i.e., in the regulation of calcium levels and calcium signaling in Purkinje cells, are the cause of several other variants of SCA. In SCA14, many of the observed mutations in the PKCγ gene were shown to increase the basal activity of PKCγ, raising the possibility that increased activity of PKCγ might be the cause of most forms of SCA14 and might also be involved in the pathogenesis of SCA in related subtypes. In this viewpoint and review article we will discuss the evidence for and against such a major role of PKCγ basal activity and will suggest a hypothesis of how PKCγ activity and the calcium signaling pathway may be involved in the pathogenesis of SCAs despite the different and sometimes opposing effects of mutations affecting these pathways. We will then widen the scope and propose a concept of SCA pathogenesis which is not primarily driven by cell death and loss of Purkinje cells but rather by dysfunction of Purkinje cells which are still present and alive in the cerebellum.
Collapse
|
10
|
Zhu X, Tan X, Wang J, Dai L, Li J, Guan X, Wang Z, Zhang M, Hu J, Bai Y, Guo H. Disruption of Intracellular Calcium Homeostasis Leads to ERLIN2-Linked Hereditary Spastic Paraplegia in Patient-Derived Stem Cell Models. Hum Mutat 2023; 2023:4834423. [PMID: 40225166 PMCID: PMC11919107 DOI: 10.1155/2023/4834423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/27/2023] [Accepted: 05/25/2023] [Indexed: 04/15/2025]
Abstract
Hereditary spastic paraplegia (HSP) is a category of neurodegenerative illnesses with significant clinical and genetic heterogeneity. Homozygous truncated variants of the ERLIN2 gene lead to HSP18 (MIM #611225). However, it is still unclear whether there is an autosomal dominant pathogenic pattern. The specific molecular mechanism needs to be investigated. We generated patient-derived iPSC models to study the mechanism of ERLIN2 heterogeneous variants leading to HSP. We identified a heterozygous missense variant p.Val71Ala of ERLIN2 in an HSP family. Based on IP-mass spectrometry, we found that the ERLIN2 heterozygous missense variant protein recruited the ubiquitin E3 ligase RNF213 to degrade IP3R1. The degradation of IP3R1 leads to the reduction of intracellular free calcium, which triggered endoplasmic reticulum (ER) stress-mediated apoptosis. Calcium homeostasis imbalance inhibited the MAPK signaling pathway that contributed to decreased cell proliferation. In summary, these results suggest that the autosomal dominant inheritance of heterozygous missense variants in ERLIN2 is a novel pathogenic mode of HSP. Furthermore, the disruption of intracellular calcium homeostasis is the pathological mechanism.
Collapse
Affiliation(s)
- Xintong Zhu
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Xiaoyin Tan
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
- NHC Key Laboratory of Birth Defects and Reproductive Health (Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute), Chongqing 400020, China
| | - Junwen Wang
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Limeng Dai
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Jia Li
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Xingying Guan
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Ziyi Wang
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Mao Zhang
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Jun Hu
- Department of Neurology, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Yun Bai
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hong Guo
- Department of Medical Genetics, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
- NHC Key Laboratory of Birth Defects and Reproductive Health (Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute), Chongqing 400020, China
- Department of Gynaecology and Obstetrics, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
11
|
Dorigo A, Valishetti K, Hetsch F, Matsui H, Meier JC, Namikawa K, Köster RW. Functional regionalization of the differentiating cerebellar Purkinje cell population occurs in an activity-dependent manner. Front Mol Neurosci 2023; 16:1166900. [PMID: 37181649 PMCID: PMC10174242 DOI: 10.3389/fnmol.2023.1166900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction The cerebellum is organized into functional regions each dedicated to process different motor or sensory inputs for controlling different locomotor behaviors. This functional regionalization is prominent in the evolutionary conserved single-cell layered Purkinje cell (PC) population. Fragmented gene expression domains suggest a genetic organization of PC layer regionalization during cerebellum development. However, the establishment of such functionally specific domains during PC differentiation remained elusive. Methods and results We show the progressive emergence of functional regionalization of PCs from broad responses to spatially restricted regions in zebrafish by means of in vivo Ca2+-imaging during stereotypic locomotive behavior. Moreover, we reveal that formation of new dendritic spines during cerebellar development using in vivo imaging parallels the time course of functional domain development. Pharmacological as well as cell-type specific optogenetic inhibition of PC neuronal activity results in reduced PC dendritic spine density and an altered stagnant pattern of functional domain formation in the PC layer. Discussion Hence, our study suggests that functional regionalization of the PC layer is driven by physiological activity of maturing PCs themselves.
Collapse
Affiliation(s)
- Alessandro Dorigo
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Komali Valishetti
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Florian Hetsch
- Cell Physiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Institute of Pathophysiology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Hideaki Matsui
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| | - Jochen C. Meier
- Cell Physiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kazuhiko Namikawa
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
- *Correspondence: Kazuhiko Namikawa,
| | - Reinhard W. Köster
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
- Reinhard W. Köster,
| |
Collapse
|
12
|
Egorova PA, Bezprozvanny IB. Electrophysiological Studies Support Utility of Positive Modulators of SK Channels for the Treatment of Spinocerebellar Ataxia Type 2. CEREBELLUM (LONDON, ENGLAND) 2022; 21:742-749. [PMID: 34978024 DOI: 10.1007/s12311-021-01349-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/18/2021] [Indexed: 06/14/2023]
Abstract
Spinocerebellar ataxia type 2 (SCA2) is an incurable hereditary disorder accompanied by cerebellar degeneration following ataxic symptoms. The causative gene for SCA2 is ATXN2. The ataxin-2 protein is involved in RNA metabolism; the polyQ expansion may interrupt ataxin-2 interaction with its molecular targets, thus representing a loss-of-function mutation. However, mutant ataxin-2 protein also displays the features of gain-of-function mutation since it forms the aggregates in SCA2 cells and also enhances the IP3-induced calcium release in affected neurons. The cerebellar Purkinje cells (PCs) are primarily affected in SCA2. Their tonic pacemaker activity is crucial for the proper cerebellar functioning. Disturbances in PC pacemaking are observed in many ataxic disorders. The abnormal intrinsic pacemaking was reported in mouse models of episodic ataxia type 2 (EA2), SCA1, SCA2, SCA3, SCA6, Huntington's disease (HD), and in some other murine models of the disorders associated with the cerebellar degeneration. In our studies using SCA2-58Q transgenic mice via cerebellar slice recording and in vivo recording from urethane-anesthetized mice and awake head-fixed mice, we have demonstrated the impaired firing frequency and irregularity of PCs in these mice. PC pacemaker activity is regulated by SK channels. The pharmacological activation of SK channels has demonstrated some promising results in the electrophysiological experiments on EA2, SCA1, SCA2, SCA3, SCA6, HD mice, and also on mutant CACNA1A mice. In our studies, we have reported that the SK activators CyPPA and NS309 converted bursting activity into tonic, while oral treatment with CyPPA and NS13001 significantly improved motor performance and PC morphology in SCA2 mice. The i.p. injections of chlorzoxazone (CHZ) during in vivo recording sessions converted bursting cells into tonic in anesthetized SCA2 mice. And, finally, long-term injections of CHZ recovered the precision of PC pacemaking activity in awake SCA2 mice and alleviated their motor decline. Thus, the SK activation can be used as a potential way to treat SCA2 and other diseases accompanied by cerebellar degeneration.
Collapse
Affiliation(s)
- Polina A Egorova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia.
| | - Ilya B Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia.
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
13
|
Ghanekar SD, Kuo SH, Staffetti JS, Zesiewicz TA. Current and Emerging Treatment Modalities for Spinocerebellar Ataxias. Expert Rev Neurother 2022; 22:101-114. [PMID: 35081319 DOI: 10.1080/14737175.2022.2029703] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Spinocerebellar ataxias (SCA) are a group of rare neurodegenerative diseases that dramatically affect the lives of affected individuals and their families. Despite having a clear understanding of SCA's etiology, there are no current symptomatic or neuroprotective treatments approved by the FDA. AREAS COVERED Research efforts have greatly expanded the possibilities for potential treatments, including both pharmacological and non-pharmacological interventions. Great attention is also being given to novel therapeutics based in gene therapy, neurostimulation, and molecular targeting. This review article will address the current advances in the treatment of SCA and what potential interventions are on the horizon. EXPERT OPINION SCA is a highly complex and multifaceted disease family with the majority of research emphasizing symptomatic pharmacologic therapies. As pre-clinical trials for SCA and clinical trials for other neurodegenerative conditions illuminate the efficacy of disease modifying therapies such as AAV-mediated gene therapy and ASOs, the potential for addressing SCA at the pre-symptomatic stage is increasingly promising.
Collapse
Affiliation(s)
- Shaila D Ghanekar
- University of South Florida (USF) Department of Neurology, USF Ataxia Research Center, Tampa, Florida, USA.,James A Haley Veteran's Hospital, Tampa, Florida, USA
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University, New York, New York, USA.,Initiative for Columbia Ataxia and Tremor, New York, New York, USA
| | - Joseph S Staffetti
- University of South Florida (USF) Department of Neurology, USF Ataxia Research Center, Tampa, Florida, USA.,James A Haley Veteran's Hospital, Tampa, Florida, USA
| | - Theresa A Zesiewicz
- University of South Florida (USF) Department of Neurology, USF Ataxia Research Center, Tampa, Florida, USA.,James A Haley Veteran's Hospital, Tampa, Florida, USA
| |
Collapse
|
14
|
Wu QW, Kapfhammer JP. Serine/threonine kinase 17b (STK17B) signalling regulates Purkinje cell dendritic development and is altered in multiple spinocerebellar ataxias. Eur J Neurosci 2021; 54:6673-6684. [PMID: 34536317 PMCID: PMC9292345 DOI: 10.1111/ejn.15465] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 11/28/2022]
Abstract
Serine/threonine kinase 17b (STK17B, also known as DRAK2) is known to be a downstream effector of protein kinase C (PKC) in the immune system, in particular T lymphocytes. PKC activity also plays a critical role for dendritic development and synaptic maturation and plasticity in cerebellar Purkinje cells. We present evidence that STK17B is strongly expressed in mouse cerebellar Purkinje cells starting in the early postnatal period and remaining highly expressed throughout adult stages and that STK17B is a target of PKC phosphorylation in the cerebellum. STK17B overexpression potentiates the morphological changes of Purkinje cells seen after PKC activation, suggesting that it is a downstream effector of PKC. A phosphorylation mimetic STK17B variant induced a marked reduction of Purkinje cell dendritic tree size, whereas the inhibition of STK17B with the novel compound 16 (Cpd16) could partially rescue the morphological changes of the Purkinje cell dendritic tree after PKC activation. These findings show that STK17B signalling is an important downstream effector of PKC activation in Purkinje cells. Furthermore, STK17B was identified as a molecule being transcriptionally downregulated in mouse models of SCA1, SCA7, SCA14 and SCA41. The reduced expression of STK17B in these mouse models might protect Purkinje cell dendrites from the negative effects of overactivated PKC signalling. Our findings provide new insights in the role of STK17B for Purkinje cell dendritic development and the pathology of SCAs.
Collapse
Affiliation(s)
- Qin-Wei Wu
- Institute of Anatomy, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Josef P Kapfhammer
- Institute of Anatomy, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
15
|
Cellular functions of the protein kinase ATM and their relevance to human disease. Nat Rev Mol Cell Biol 2021; 22:796-814. [PMID: 34429537 DOI: 10.1038/s41580-021-00394-2] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
The protein kinase ataxia telangiectasia mutated (ATM) is a master regulator of double-strand DNA break (DSB) signalling and stress responses. For three decades, ATM has been investigated extensively to elucidate its roles in the DNA damage response (DDR) and in the pathogenesis of ataxia telangiectasia (A-T), a human neurodegenerative disease caused by loss of ATM. Although hundreds of proteins have been identified as ATM phosphorylation targets and many important roles for this kinase have been identified, it is still unclear how ATM deficiency leads to the early-onset cerebellar degeneration that is common in all individuals with A-T. Recent studies suggest the existence of links between ATM deficiency and other cerebellum-specific neurological disorders, as well as the existence of broader similarities with more common neurodegenerative disorders. In this Review, we discuss recent structural insights into ATM regulation, and possible aetiologies of A-T phenotypes, including reactive oxygen species, mitochondrial dysfunction, alterations in transcription, R-loop metabolism and alternative splicing, defects in cellular proteostasis and metabolism, and potential pathogenic roles for hyper-poly(ADP-ribosyl)ation.
Collapse
|
16
|
Huang W, Dai M, Qiu T, Liang T, Xie J, Mi C, Zhao J, Chen W, Tian P, Zhang S, Zhang H. Novel lncRNA-HZ04 promotes BPDE-induced human trophoblast cell apoptosis and miscarriage by upregulating IP 3 R 1 /CaMKII/SGCB pathway by competitively binding with miR-hz04. FASEB J 2021; 35:e21789. [PMID: 34383983 DOI: 10.1096/fj.202100376rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/09/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022]
Abstract
Normal pregnancy is essential for human reproduction. However, BaP (benzo(a)pyrene) and its metabolite BPDE (benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide) could cause dysfunctions of human trophoblast cells and might further induce miscarriage. Yet, the underlying mechanisms remain largely unknown. Herein, we identified a novel upregulated lnc-HZ04 and a novel downregulated miR-hz04 in villous tissues of unexplained recurrent miscarriage (RM) relative to those in healthy control tissues and also in BPDE-treated human trophoblast cells. Lnc-HZ04 directly and specifically bound with miR-hz04, diminished the reduction effects of miR-hz04 on IP3 R1 mRNA expression level and on IP3 R1 mRNA stability, and then activated the Ca2+ -mediated IP3 R1 /p-CaMKII/SGCB pathway, which further promoted trophoblast cell apoptosis. The miR-hz04 target site on lnc-HZ04 played crucial roles in these regulations. In normal trophoblast, relatively less lnc-HZ04 and more miR-hz04 suppressed this apoptosis pathway and gave normal pregnancy. After exposure to BPDE or in RM tissues, p53 was upregulated, which might promote p53-mediated lnc-HZ04 transcription. Relatively more lnc-HZ04 and less miR-hz04 activated this apoptosis pathway and might further induce miscarriage. BaP could also induce mice miscarriage by upregulating its corresponding murine apoptosis pathway. Therefore, BPDE-induced apoptosis of human trophoblast cells was associated with the occurrence of miscarriage. This work discovered the regulation roles of lnc-HZ04 and miR-hz04 and provided scientific and clinical understanding of the occurrence of unexplained miscarriage.
Collapse
Affiliation(s)
- Wenxin Huang
- Research Center for Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Mengyuan Dai
- Research Center for Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Taotao Qiu
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tingting Liang
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayu Xie
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chenyang Mi
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jingsong Zhao
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Weina Chen
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Peng Tian
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Shuming Zhang
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huidong Zhang
- Research Center for Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Purkinje Neurons with Loss of STIM1 Exhibit Age-Dependent Changes in Gene Expression and Synaptic Components. J Neurosci 2021; 41:3777-3798. [PMID: 33737457 DOI: 10.1523/jneurosci.2401-20.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
The stromal interaction molecule 1 (STIM1) is an ER-Ca2+ sensor and an essential component of ER-Ca2+ store operated Ca2+ entry. Loss of STIM1 affects metabotropic glutamate receptor 1 (mGluR1)-mediated synaptic transmission, neuronal Ca2+ homeostasis, and intrinsic plasticity in Purkinje neurons (PNs). Long-term changes of intracellular Ca2+ signaling in PNs led to neurodegenerative conditions, as evident in individuals with mutations of the ER-Ca2+ channel, the inositol 1,4,5-triphosphate receptor. Here, we asked whether changes in such intrinsic neuronal properties, because of loss of STIM1, have an age-dependent impact on PNs. Consequently, we analyzed mRNA expression profiles and cerebellar morphology in PN-specific STIM1 KO mice (STIM1PKO ) of both sexes across ages. Our study identified a requirement for STIM1-mediated Ca2+ signaling in maintaining the expression of genes belonging to key biological networks of synaptic function and neurite development among others. Gene expression changes correlated with altered patterns of dendritic morphology and greater innervation of PN dendrites by climbing fibers, in aging STIM1PKO mice. Together, our data identify STIM1 as an important regulator of Ca2+ homeostasis and neuronal excitability in turn required for maintaining the optimal transcriptional profile of PNs with age. Our findings are significant in the context of understanding how dysregulated calcium signals impact cellular mechanisms in multiple neurodegenerative disorders.SIGNIFICANCE STATEMENT In Purkinje neurons (PNs), the stromal interaction molecule 1 (STIM1) is required for mGluR1-dependent synaptic transmission, refilling of ER Ca2+ stores, regulation of spike frequency, and cerebellar memory consolidation. Here, we provide evidence for a novel role of STIM1 in maintaining the gene expression profile and optimal synaptic connectivity of PNs. Expression of genes related to neurite development and synaptic organization networks is altered in PNs with persistent loss of STIM1. In agreement with these findings the dendritic morphology of PNs and climbing fiber innervations on PNs also undergo significant changes with age. These findings identify a new role for dysregulated intracellular calcium signaling in neurodegenerative disorders and provide novel therapeutic insights.
Collapse
|
18
|
Yeo AJ, Chong KL, Gatei M, Zou D, Stewart R, Withey S, Wolvetang E, Parton RG, Brown AD, Kastan MB, Coman D, Lavin MF. Impaired endoplasmic reticulum-mitochondrial signaling in ataxia-telangiectasia. iScience 2021; 24:101972. [PMID: 33437944 PMCID: PMC7788243 DOI: 10.1016/j.isci.2020.101972] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 12/21/2022] Open
Abstract
There is evidence that ATM mutated in ataxia-telangiectasia (A-T) plays a key role in protecting against mitochondrial dysfunction, the mechanism for which remains unresolved. We demonstrate here that ATM-deficient cells are exquisitely sensitive to nutrient deprivation, which can be explained by defective cross talk between the endoplasmic reticulum (ER) and the mitochondrion. Tethering between these two organelles in response to stress was reduced in cells lacking ATM, and consistent with this, Ca2+ release and transfer between ER and mitochondria was reduced dramatically when compared with control cells. The impact of this on mitochondrial function was evident from an increase in oxygen consumption rates and a defect in mitophagy in ATM-deficient cells. Our findings reveal that ER-mitochondrial connectivity through IP3R1-GRP75-VDAC1, to maintain Ca2+ homeostasis, as well as an abnormality in mitochondrial fusion defective in response to nutrient stress, can account for at least part of the mitochondrial dysfunction observed in A-T cells.
Collapse
Affiliation(s)
- Abrey J. Yeo
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| | - Kok L. Chong
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| | - Magtouf Gatei
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| | - Dongxiu Zou
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| | | | - Sarah Withey
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, Australia
| | - Ernst Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, St Lucia, Brisbane, Australia
| | | | | | - David Coman
- Queensland Children's Hospital, Brisbane, Australia
| | - Martin F. Lavin
- University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, Australia
| |
Collapse
|
19
|
Wu QW, Kapfhammer JP. Modulation of Increased mGluR1 Signaling by RGS8 Protects Purkinje Cells From Dendritic Reduction and Could Be a Common Mechanism in Diverse Forms of Spinocerebellar Ataxia. Front Cell Dev Biol 2021; 8:569889. [PMID: 33553137 PMCID: PMC7858651 DOI: 10.3389/fcell.2020.569889] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Spinocerebellar ataxias (SCAs) are a group of hereditary neurodegenerative diseases which are caused by diverse genetic mutations in a variety of different genes. We have identified RGS8, a regulator of G-protein signaling, as one of the genes which are dysregulated in different mouse models of SCA (e.g., SCA1, SCA2, SCA7, and SCA14). In the moment, little is known about the role of RGS8 for pathogenesis of spinocerebellar ataxia. We have studied the expression of RGS8 in the cerebellum in more detail and show that it is specifically expressed in mouse cerebellar Purkinje cells. In a mouse model of SCA14 with increased PKCγ activity, RGS8 expression was also increased. RGS8 overexpression could partially counteract the negative effects of DHPG-induced mGluR1 signaling for the expansion of Purkinje cell dendrites. Our results suggest that the increased expression of RGS8 is an important mediator of mGluR1 pathway dysregulation in Purkinje cells. These findings provide new insights in the role of RGS8 and mGluR1 signaling in Purkinje cells and for the pathology of SCAs.
Collapse
Affiliation(s)
- Qin-Wei Wu
- Institute of Anatomy, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Josef P Kapfhammer
- Institute of Anatomy, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
20
|
A New Mouse Model Related to SCA14 Carrying a Pseudosubstrate Domain Mutation in PKCγ Shows Perturbed Purkinje Cell Maturation and Ataxic Motor Behavior. J Neurosci 2021; 41:2053-2068. [PMID: 33478986 PMCID: PMC7939089 DOI: 10.1523/jneurosci.1946-20.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 01/15/2023] Open
Abstract
Spinocerebellar ataxias (SCAs) are diseases characterized by cerebellar atrophy and loss of Purkinje neurons caused by mutations in diverse genes. In SCA14, the disease is caused by point mutations or small deletions in protein kinase C γ (PKCγ), a crucial signaling protein in Purkinje cells. It is still unclear whether increased or decreased PKCγ activity may be involved in the SCA14 pathogenesis. In this study, we present a new knock-in mouse model related to SCA14 with a point mutation in the pseudosubstrate domain, PKCγ-A24E, known to induce a constitutive PKCγ activation. In this protein conformation, the kinase domain of PKCγ is activated, but at the same time the protein is subject to dephosphorylation and protein degradation. As a result, we find a dramatic reduction of PKCγ protein expression in PKCγ-A24E mice of either sex. Despite this reduction, there is clear evidence for an increased PKC activity in Purkinje cells from PKCγ-A24E mice. Purkinje cells derived from PKCγ-A24E have short thickened dendrites typical for PKC activation. These mice also develop a marked ataxia and signs of Purkinje cell dysfunction making them an interesting new mouse model related to SCA. Recently, a similar mutation in a human patient was discovered and found to be associated with overt SCA14. RNA profiling of PKCγ-A24E mice showed a dysregulation of related signaling pathways, such as mGluR1 or mTOR. Our results show that the induction of PKCγ activation in Purkinje cells results in the SCA-like phenotype indicating PKC activation as one pathogenetic avenue leading to a SCA.SIGNIFICANCE STATEMENT Spinocerebellar ataxias (SCAs) are hereditary diseases affecting cerebellar Purkinje cells and are a one of neurodegenerative diseases. While mutation in several genes have been identified as causing SCAs, it is unclear how these mutations cause the disease phenotype. Mutations in PKCγ cause one subtype of SCAs, SCA14. In this study, we have generated a knock-in mouse with a mutation in the pseudosubstrate domain of PKCγ, which keeps PKCγ in the constitutive active open conformation. We show that this mutation leading to a constant activation of PKCγ results in a SCA-like phenotype in these mice. Our findings establish the constant activation of PKC signaling as one pathogenetic avenue leading to an SCA phenotype and a mechanism causing a neurodegenerative disease.
Collapse
|
21
|
de Oude NL, Hoebeek FE, Ten Brinke MM, de Zeeuw CI, Boele HJ. Pavlovian eyeblink conditioning is severely impaired in tottering mice. J Neurophysiol 2020; 125:398-407. [PMID: 33326350 DOI: 10.1152/jn.00578.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cacna1a encodes the pore-forming α1A subunit of CaV2.1 voltage-dependent calcium channels, which regulate neuronal excitability and synaptic transmission. Purkinje cells in the cortex of cerebellum abundantly express these CaV2.1 channels. Here, we show that homozygous tottering (tg) mice, which carry a loss-of-function Cacna1a mutation, exhibit severely impaired learning in Pavlovian eyeblink conditioning, which is a cerebellar-dependent learning task. Performance of reflexive eyeblinks is unaffected in tg mice. Transient seizure activity in tg mice further corrupted the amplitude of eyeblink conditioned responses. Our results indicate that normal calcium homeostasis is imperative for cerebellar learning and that the oscillatory state of the brain can affect the expression thereof.NEW & NOTEWORTHY In this study, we confirm the importance of normal calcium homeostasis in neurons for learning and memory formation. In a mouse model with a mutation in an essential calcium channel that is abundantly expressed in the cerebellum, we found severely impaired learning in eyeblink conditioning. Eyeblink conditioning is a cerebellar-dependent learning task. During brief periods of brain-wide oscillatory activity, as a result of the mutation, the expression of conditioned eyeblinks was even further disrupted.
Collapse
Affiliation(s)
- Nina L de Oude
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Freek E Hoebeek
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands.,Department for Developmental Origins of Disease, Wilhelmina Children's Hospital, Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Chris I de Zeeuw
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands.,Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Henk-Jan Boele
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands.,Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| |
Collapse
|
22
|
Spinocerebellar ataxia type 23 (SCA23): a review. J Neurol 2020; 268:4630-4645. [PMID: 33175256 DOI: 10.1007/s00415-020-10297-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 02/08/2023]
Abstract
Spinocerebellar ataxias (SCAs), formerly known as autosomal dominant cerebellar ataxias (ADCAs), are a group of hereditary heterogeneous neurodegenerative diseases. Gait, progressive ataxia, dysarthria, and eye movement disorder are common symptoms of spinocerebellar ataxias. Other symptoms include peripheral neuropathy, cognitive impairment, psychosis, and seizures. Patients may lose their lives due to out of coordinated respiration and/or swallowing. Neurological signs cover pyramidal or extrapyramidal signs, spasm, ophthalmoplegia, hyperactive deep tendon reflexes, and so on. Different subtypes of SCAs present various clinical features. Spinocerebellar ataxia type 23 (SCA23), one subtype of the SCA family, is characterized by mutant prodynorphin (PDYN) gene. Based on literatures, this review details a series of SCA23, to improve a whole understanding of clinicians and point out the potential research direction of this dysfunction, including a history, pathophysiological mechanism, diagnosis and differential diagnosis, epigenetics, penetrance and prevalence, genetic counseling, treatment and prognosis.
Collapse
|
23
|
Neves-Carvalho A, Duarte-Silva S, Teixeira-Castro A, Maciel P. Polyglutamine spinocerebellar ataxias: emerging therapeutic targets. Expert Opin Ther Targets 2020; 24:1099-1119. [PMID: 32962458 DOI: 10.1080/14728222.2020.1827394] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Six of the most frequent dominantly inherited spinocerebellar ataxias (SCAs) worldwide - SCA1, SCA2, SCA3, SCA6, SCA7, and SCA17 - are caused by an expansion of a polyglutamine (polyQ) tract in the corresponding proteins. While the identification of the causative mutation has advanced knowledge on the pathogenesis of polyQ SCAs, effective therapeutics able to mitigate the severe clinical manifestation of these highly incapacitating disorders are not yet available. AREAS COVERED This review provides a comprehensive and critical perspective on well-established and emerging therapeutic targets for polyQ SCAs; it aims to inspire prospective drug discovery efforts. EXPERT OPINION The landscape of polyQ SCAs therapeutic targets and strategies includes (1) the mutant genes and proteins themselves, (2) enhancement of endogenous protein quality control responses, (3) abnormal protein-protein interactions of the mutant proteins, (4) disturbed neuronal function, (5) mitochondrial function, energy availability and oxidative stress, and (6) glial dysfunction, growth factor or hormone imbalances. Challenges include gaining a clearer definition of therapeutic targets for the drugs in clinical development, the discovery of novel drug-like molecules for challenging key targets, and the attainment of a stronger translation of preclinical findings to the clinic.
Collapse
Affiliation(s)
- Andreia Neves-Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho , Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory , Braga, Guimarães, Portugal
| |
Collapse
|
24
|
Orem BC, Rajaee A, Stirling DP. IP 3R-mediated intra-axonal Ca 2+ release contributes to secondary axonal degeneration following contusive spinal cord injury. Neurobiol Dis 2020; 146:105123. [PMID: 33011333 DOI: 10.1016/j.nbd.2020.105123] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/15/2020] [Accepted: 09/28/2020] [Indexed: 01/11/2023] Open
Abstract
Secondary axonal loss contributes to the persistent functional disability following trauma. Consequently, preserving axons following spinal cord injury (SCI) is a major therapeutic goal to improve neurological outcome; however, the complex molecular mechanisms that mediate secondary axonal degeneration remain unclear. We previously showed that IP3R-mediated Ca2+ release contributes to axonal dieback and axonal loss following an ex vivo laser-induced SCI. Nevertheless, targeting IP3R in a clinically relevant in vivo model of SCI and determining its contribution to secondary axonal degeneration has yet to be explored. Here we used intravital two-photon excitation microscopy to assess the role of IP3R in secondary axonal degeneration in real-time after a contusive-SCI in vivo. To visualize Ca2+ changes specifically in spinal axons over time, adult 6-8 week-old triple transgenic Avil-Cre:Ai9:Ai95 (sensory neuron-specific expression of tdTomato and the genetic calcium indicator GCaMP6f) mice were subjected to a mild (30 kdyn) T12 contusive-SCI and received delayed treatment with the IP3R blocker 2-APB (100 μM, intrathecal delivery at 3, and 24 h following injury) or vehicle control. To determine the IP3R subtype involved, we knocked-down IP3R3 using capped phosphodiester oligonucleotides. Delayed treatment with 2-APB significantly reduced axonal spheroids, increased axonal survival, and reduced intra-axonal Ca2+ accumulation within dorsal column axons at 24 h following SCI in vivo. Additionally, knockdown of IP3R3 yielded increased axon survival 24 h post-SCI. These results suggest that IP3R-mediated Ca2+ release contributes to secondary axonal degeneration in vivo following SCI.
Collapse
Affiliation(s)
- Ben C Orem
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY 40202, USA; Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY 40202, USA
| | - Arezoo Rajaee
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY 40202, USA; Departments of Neurological Surgery, University of Louisville, School of Medicine, Louisville, KY 40202, USA
| | - David P Stirling
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY 40202, USA; Departments of Neurological Surgery, University of Louisville, School of Medicine, Louisville, KY 40202, USA; Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY 40202, USA; Microbiology and Immunology, University of Louisville, School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
25
|
Kim J, Kim K, Mo JS, Lee Y. Atm deficiency in the DNA polymerase β null cerebellum results in cerebellar ataxia and Itpr1 reduction associated with alteration of cytosine methylation. Nucleic Acids Res 2020; 48:3678-3691. [PMID: 32123907 PMCID: PMC7144915 DOI: 10.1093/nar/gkaa140] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 02/20/2020] [Accepted: 02/27/2020] [Indexed: 12/13/2022] Open
Abstract
Genomic instability resulting from defective DNA damage responses or repair causes several abnormalities, including progressive cerebellar ataxia, for which the molecular mechanisms are not well understood. Here, we report a new murine model of cerebellar ataxia resulting from concomitant inactivation of POLB and ATM. POLB is one of key enzymes for the repair of damaged or chemically modified bases, including methylated cytosine, but selective inactivation of Polb during neurogenesis affects only a subpopulation of cortical interneurons despite the accumulation of DNA damage throughout the brain. However, dual inactivation of Polb and Atm resulted in ataxia without significant neuropathological defects in the cerebellum. ATM is a protein kinase that responds to DNA strand breaks, and mutations in ATM are responsible for Ataxia Telangiectasia, which is characterized by progressive cerebellar ataxia. In the cerebella of mice deficient for both Polb and Atm, the most downregulated gene was Itpr1, likely because of misregulated DNA methylation cycle. ITPR1 is known to mediate calcium homeostasis, and ITPR1 mutations result in genetic diseases with cerebellar ataxia. Our data suggest that dysregulation of ITPR1 in the cerebellum could be one of contributing factors to progressive ataxia observed in human genomic instability syndromes.
Collapse
Affiliation(s)
- Jusik Kim
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, The Graduate School of Ajou University, Suwon 16499, Korea
| | - Keeeun Kim
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, The Graduate School of Ajou University, Suwon 16499, Korea
| | - Jung-Soon Mo
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, The Graduate School of Ajou University, Suwon 16499, Korea
| | - Youngsoo Lee
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, The Graduate School of Ajou University, Suwon 16499, Korea
| |
Collapse
|
26
|
Chen YS, Hong ZX, Lin SZ, Harn HJ. Identifying Therapeutic Targets for Spinocerebellar Ataxia Type 3/Machado-Joseph Disease through Integration of Pathological Biomarkers and Therapeutic Strategies. Int J Mol Sci 2020; 21:ijms21093063. [PMID: 32357546 PMCID: PMC7246822 DOI: 10.3390/ijms21093063] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
Spinocerebellar ataxia type 3/Machado-Joseph disease (SCA3/MJD) is a progressive motor disease with no broadly effective treatment. However, most current therapies are based on symptoms rather than the underlying disease mechanisms. In this review, we describe potential therapeutic strategies based on known pathological biomarkers and related pathogenic processes. The three major conclusions from the current studies are summarized as follows: (i) for the drugs currently being tested in clinical trials; a weak connection was observed between drugs and SCA3/MJD biomarkers. The only two exceptions are the drugs suppressing glutamate-induced calcium influx and chemical chaperon. (ii) For most of the drugs that have been tested in animal studies, there is a direct association with pathological biomarkers. We further found that many drugs are associated with inducing autophagy, which is supported by the evidence of deficient autophagy biomarkers in SCA3/MJD, and that there may be more promising therapeutics. (iii) Some reported biomarkers lack relatively targeted drugs. Low glucose utilization, altered amino acid metabolism, and deficient insulin signaling are all implicated in SCA3/MJD, but there have been few studies on treatment strategies targeting these abnormalities. Therapeutic strategies targeting multiple pathological SCA3/MJD biomarkers may effectively block disease progression and preserve neurological function.
Collapse
Affiliation(s)
- Yu-Shuan Chen
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Correspondence: (Y.-S.C.); (H.-J.H.); Tel.: +886-3-856-1825 (Y.-S.C. & H.-J.H.); Fax: +886-3-856-0977 (H.-J.H.)
| | - Zhen-Xiang Hong
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
| | - Horng-Jyh Harn
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Department of Pathology, Hualien Tzu Chi Hospital, Tzu Chi University, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Correspondence: (Y.-S.C.); (H.-J.H.); Tel.: +886-3-856-1825 (Y.-S.C. & H.-J.H.); Fax: +886-3-856-0977 (H.-J.H.)
| |
Collapse
|
27
|
Sanyanga TA, Tastan Bishop Ö. Structural Characterization of Carbonic Anhydrase VIII and Effects of Missense Single Nucleotide Variations to Protein Structure and Function. Int J Mol Sci 2020; 21:E2764. [PMID: 32316137 PMCID: PMC7215520 DOI: 10.3390/ijms21082764] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
Abstract
Human carbonic anhydrase 8 (CA-VIII) is an acatalytic isoform of the α -CA family. Though the protein cannot hydrate CO2, CA-VIII is essential for calcium (Ca2+) homeostasis within the body, and achieves this by allosterically inhibiting the binding of inositol 1,4,5-triphosphate (IP3) to the IP3 receptor type 1 (ITPR1) protein. However, the mechanism of interaction of CA-VIII to ITPR1 is not well understood. In addition, functional defects to CA-VIII due to non-synonymous single nucleotide polymorphisms (nsSNVs) result in Ca2+ dysregulation and the development of the phenotypes such as cerebellar ataxia, mental retardation and disequilibrium syndrome 3 (CAMRQ3). The pathogenesis of CAMRQ3 is also not well understood. The structure and function of CA-VIII was characterised, and pathogenesis of CAMRQ3 investigated. Structural and functional characterisation of CA-VIII was conducted through SiteMap and CPORT to identify potential binding site residues. The effects of four pathogenic nsSNVs, S100A, S100P, G162R and R237Q, and two benign S100L and E109D variants on CA-VIII structure and function was then investigated using molecular dynamics (MD) simulations, dynamic cross correlation (DCC) and dynamic residue network (DRN) analysis. SiteMap and CPORT analyses identified 38 unique CA-VIII residues that could potentially bind to ITPR1. MD analysis revealed less conformational sampling within the variant proteins and highlighted potential increases to variant protein rigidity. Dynamic cross correlation (DCC) showed that wild-type (WT) protein residue motion is predominately anti-correlated, with variant proteins showing no correlation to greater residue correlation. DRN revealed variant-associated increases to the accessibility of the N-terminal binding site residues, which could have implications for associations with ITPR1, and further highlighted differences to the mechanism of benign and pathogenic variants. SNV presence is associated with a reduction to the usage of Trp37 in all variants, which has implications for CA-VIII stability. The differences to variant mechanisms can be further investigated to understand pathogenesis of CAMRQ3, enhancing precision medicine-related studies into CA-VIII.
Collapse
MESH Headings
- Binding Sites
- Biomarkers, Tumor/chemistry
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cerebellar Ataxia/genetics
- Cerebellar Ataxia/pathology
- Databases, Genetic
- Humans
- Inositol 1,4,5-Trisphosphate Receptors/chemistry
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Intellectual Disability/genetics
- Intellectual Disability/pathology
- Molecular Dynamics Simulation
- Mutation, Missense
- Polymorphism, Single Nucleotide
- Protein Binding
- Protein Interaction Maps
- Protein Stability
- Protein Structure, Tertiary
Collapse
Affiliation(s)
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown 6140, South Africa;
| |
Collapse
|
28
|
Bankers L, Miller C, Liu G, Thongkittidilok C, Morrison J, Poeschla EM. Development of IFN-Stimulated Gene Expression from Embryogenesis through Adulthood, with and without Constitutive MDA5 Pathway Activation. THE JOURNAL OF IMMUNOLOGY 2020; 204:2791-2807. [PMID: 32277054 DOI: 10.4049/jimmunol.1901421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/26/2020] [Indexed: 11/19/2022]
Abstract
Pathogen-associated molecular patterns (e.g., dsRNA) activate expression of IFN-stimulated genes (ISGs), which protect hosts from infection. Although transient ISG upregulation is essential for effective innate immunity, constitutive activation typically causes harmful autoimmunity in mice and humans, often including severe developmental abnormalities. We have shown that transgenic mice expressing a picornavirus RNA-dependent RNA polymerase (RdRP) outside the viral context (RdRP mice) exhibit constitutive, MDA5-dependent, and quantitatively dramatic upregulation of many ISGs, which confers broad viral infection resistance. Remarkably, RdRP mice never develop autoinflammation, interferonopathy, or other discernible abnormalities. In this study, we used RNA sequencing and other methods to analyze ISG expression across five time points from fetal development to adulthood in wild-type and RdRP mice. In RdRP mice, the proportion of upregulated ISGs increased during development, with the most dramatic induction occurring 2 wk postnatally. The amplified ISG profile is then maintained lifelong. Molecular pathways and biological functions associated with innate immune and IFN signaling are only activated postnatally, suggesting constrained fetal responsiveness to innate immune stimuli. Biological functions supporting replication of viruses are only inhibited postnatally. We further determined that the RdRP is expressed at low levels and that blocking Ifnar1 reverses the amplified ISG transcriptome in adults. In conclusion, the upregulated ISG profile of RdRP mice is mostly triggered early postnatally, is maintained through adulthood, and requires ongoing type I IFN signaling to maintain it. The model provides opportunities to study the systems biology of innate immunity and to determine how sustained ISG upregulation can be compatible with robust health.
Collapse
Affiliation(s)
- Laura Bankers
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Caitlin Miller
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Guoqi Liu
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Chommanart Thongkittidilok
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - James Morrison
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| | - Eric M Poeschla
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver School of Medicine, Aurora, CO 80045
| |
Collapse
|
29
|
Liu Q, Huang S, Yin P, Yang S, Zhang J, Jing L, Cheng S, Tang B, Li XJ, Pan Y, Li S. Cerebellum-enriched protein INPP5A contributes to selective neuropathology in mouse model of spinocerebellar ataxias type 17. Nat Commun 2020; 11:1101. [PMID: 32107387 PMCID: PMC7046734 DOI: 10.1038/s41467-020-14931-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 02/11/2020] [Indexed: 01/01/2023] Open
Abstract
Spinocerebellar ataxias 17 (SCA17) is caused by polyglutamine (polyQ) expansion in the TATA box-binding protein (TBP). The selective neurodegeneration in the cerebellum in SCA17 raises the question of why ubiquitously expressed polyQ proteins can cause neurodegeneration in distinct brain regions in different polyQ diseases. By expressing mutant TBP in different brain regions in adult wild-type mice via stereotaxic injection of adeno-associated virus, we found that adult cerebellar neurons are particularly vulnerable to mutant TBP. In SCA17 knock-in mice, mutant TBP inhibits SP1-mediated gene transcription to down-regulate INPP5A, a protein that is highly abundant in the cerebellum. CRISPR/Cas9-mediated deletion of Inpp5a in the cerebellum of wild-type mice leads to Purkinje cell degeneration, and Inpp5a overexpression decreases inositol 1,4,5-trisphosphate (IP3) levels and ameliorates Purkinje cell degeneration in SCA17 knock-in mice. Our findings demonstrate the important contribution of a tissue-specific protein to the polyQ protein-mediated selective neuropathology.
Collapse
Affiliation(s)
- Qiong Liu
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shanshan Huang
- Department of Neurology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Yin
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Su Yang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Jennifer Zhang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Liang Jing
- Department of Emergency, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Siying Cheng
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Beisha Tang
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xiao-Jiang Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Yongcheng Pan
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Shihua Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China.
| |
Collapse
|
30
|
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: 27] [Impact Index Per Article: 4.5] [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
|
31
|
Sen NE, Arsovic A, Meierhofer D, Brodesser S, Oberschmidt C, Canet-Pons J, Kaya ZE, Halbach MV, Gispert S, Sandhoff K, Auburger G. In Human and Mouse Spino-Cerebellar Tissue, Ataxin-2 Expansion Affects Ceramide-Sphingomyelin Metabolism. Int J Mol Sci 2019; 20:E5854. [PMID: 31766565 PMCID: PMC6928749 DOI: 10.3390/ijms20235854] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023] Open
Abstract
Ataxin-2 (human gene symbol ATXN2) acts during stress responses, modulating mRNA translation and nutrient metabolism. Ataxin-2 knockout mice exhibit progressive obesity, dyslipidemia, and insulin resistance. Conversely, the progressive ATXN2 gain of function due to the fact of polyglutamine (polyQ) expansions leads to a dominantly inherited neurodegenerative process named spinocerebellar ataxia type 2 (SCA2) with early adipose tissue loss and late muscle atrophy. We tried to understand lipid dysregulation in a SCA2 patient brain and in an authentic mouse model. Thin layer chromatography of a patient cerebellum was compared to the lipid metabolome of Atxn2-CAG100-Knockin (KIN) mouse spinocerebellar tissue. The human pathology caused deficits of sulfatide, galactosylceramide, cholesterol, C22/24-sphingomyelin, and gangliosides GM1a/GD1b despite quite normal levels of C18-sphingomyelin. Cerebellum and spinal cord from the KIN mouse showed a consistent decrease of various ceramides with a significant elevation of sphingosine in the more severely affected spinal cord. Deficiency of C24/26-sphingomyelins contrasted with excess C18/20-sphingomyelin. Spinocerebellar expression profiling revealed consistent reductions of CERS protein isoforms, Sptlc2 and Smpd3, but upregulation of Cers2 mRNA, as prominent anomalies in the ceramide-sphingosine metabolism. Reduction of Asah2 mRNA correlated to deficient S1P levels. In addition, downregulations for the elongase Elovl1, Elovl4, Elovl5 mRNAs and ELOVL4 protein explain the deficit of very long-chain sphingomyelin. Reduced ASMase protein levels correlated to the accumulation of long-chain sphingomyelin. Overall, a deficit of myelin lipids was prominent in SCA2 nervous tissue at prefinal stage and not compensated by transcriptional adaptation of several metabolic enzymes. Myelination is controlled by mTORC1 signals; thus, our human and murine observations are in agreement with the known role of ATXN2 yeast, nematode, and mouse orthologs as mTORC1 inhibitors and autophagy promoters.
Collapse
Affiliation(s)
- Nesli-Ece Sen
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
- Faculty of Biosciences, Goethe-University, 60438 Frankfurt am Main, Germany
| | - Aleksandar Arsovic
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany;
| | - Susanne Brodesser
- Membrane Biology and Lipid Biochemistry Unit, Life and Medical Sciences Institute, University of Bonn, 53121 Bonn, Germany;
| | - Carola Oberschmidt
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Júlia Canet-Pons
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Zeynep-Ece Kaya
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
- Cerrahpasa School of Medicine, Istanbul University, 34098 Istanbul, Turkey
| | - Melanie-Vanessa Halbach
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Suzana Gispert
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| | - Konrad Sandhoff
- Membrane Biology and Lipid Biochemistry Unit, Life and Medical Sciences Institute, University of Bonn, 53121 Bonn, Germany;
| | - Georg Auburger
- Experimental Neurology, Building 89, Goethe University Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany; (N.-E.S.); (A.A.); (C.O.); (J.C.-P.); (Z.-E.K.); (M.-V.H.); (S.G.)
| |
Collapse
|
32
|
Egorova PA, Bezprozvanny IB. Molecular Mechanisms and Therapeutics for Spinocerebellar Ataxia Type 2. Neurotherapeutics 2019; 16:1050-1073. [PMID: 31435879 PMCID: PMC6985344 DOI: 10.1007/s13311-019-00777-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The effective therapeutic treatment and the disease-modifying therapy for spinocerebellar ataxia type 2 (SCA2) (a progressive hereditary disease caused by an expansion of polyglutamine in the ataxin-2 protein) is not available yet. At present, only symptomatic treatment and methods of palliative care are prescribed to the patients. Many attempts were made to study the physiological, molecular, and biochemical changes in SCA2 patients and in a variety of the model systems to find new therapeutic targets for SCA2 treatment. A better understanding of the uncovered molecular mechanisms of the disease allowed the scientific community to develop strategies of potential therapy and helped to create some promising therapeutic approaches for SCA2 treatment. Recent progress in this field will be discussed in this review article.
Collapse
Affiliation(s)
- Polina A Egorova
- Laboratory of Molecular Neurodegeneration, Peter the Great St.Petersburg Polytechnic University, St. Petersburg, 195251, Russia
| | - Ilya B Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St.Petersburg Polytechnic University, St. Petersburg, 195251, Russia.
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, ND12.200, Dallas, Texas, 75390, USA.
| |
Collapse
|
33
|
Synofzik M, Puccio H, Mochel F, Schöls L. Autosomal Recessive Cerebellar Ataxias: Paving the Way toward Targeted Molecular Therapies. Neuron 2019; 101:560-583. [PMID: 30790538 DOI: 10.1016/j.neuron.2019.01.049] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/20/2018] [Accepted: 01/23/2019] [Indexed: 12/22/2022]
Abstract
Autosomal-recessive cerebellar ataxias (ARCAs) comprise a heterogeneous group of rare degenerative and metabolic genetic diseases that share the hallmark of progressive damage of the cerebellum and its associated tracts. This Review focuses on recent translational research in ARCAs and illustrates the steps from genetic characterization to preclinical and clinical trials. The emerging common pathways underlying ARCAs include three main clusters: mitochondrial dysfunction, impaired DNA repair, and complex lipid homeostasis. Novel ARCA treatments might target common hubs in pathogenesis by modulation of gene expression, stem cell transplantation, viral gene transfer, or interventions in faulty pathways. All these translational steps are addressed in current ARCA research, leading to the expectation that novel treatments for ARCAs will be reached in the next decade.
Collapse
Affiliation(s)
- Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Hélène Puccio
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67404 Illkirch, France; INSERM, U1258, 67404 Illkirch, France; CNRS, UMR7104, 67404 Illkirch, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Fanny Mochel
- Sorbonne Université, UPMC-Paris 6, UMR S 1127 and Inserm U 1127, and CNRS UMR 7225, and Institut du Cerveau et de la Moelle épinière, 75013 Paris, France; Department of Genetics and Reference Centre for Adult Neurometabolic Diseases, AP-HP, La Pitié-Salpêtriere University Hospital, Paris, France
| | - Ludger Schöls
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| |
Collapse
|
34
|
Park SY, Yoo YM, Jung EM, Jeung EB. Distribution of and steroid hormone effects on calbindin-D 9k in the immature rat brain. Brain Res Bull 2019; 152:225-235. [PMID: 31357009 DOI: 10.1016/j.brainresbull.2019.07.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/08/2019] [Accepted: 07/23/2019] [Indexed: 01/06/2023]
Abstract
Calbindin-D9k (CaBP-9k), one of the major calcium-binding and calcium-buffering proteins, is important in the physiological functioning of organs. The neuroanatomical localization of CaBP-9k in the rodent brain has not been reported; thus, this study investigated the neuroanatomical distribution of CaBP-9k and the regulation of CaBP-9k expression on steroid hormones in the immature rat brain. To confirm the influence of steroid hormones on CaBP-9k expression, immature female rats were injected for 5 days with estrogen (E2), progesterone (P4), dexamethasone (DEX), and their antagonists (ICI 182, 780 and RU 486). The localization and expression of the CaBP-9k protein in brain regions were identified by immunofluorescence and western blot assays, respectively. We observed that CaBP-9k expression was especially strong in hypothalamus, cerebellum, and brain stem. In addition, CaBP-9k was colocalized with mature-, GABAergic, dopaminergic, and oxytocinergic neurons. We also observed that the CaBP-9k protein level was significantly increased by P4 and reversed by antagonist RU 486 treatment in immature rat brain. In summary, CaBP-9k positive cells have a wide distribution in the immature rat brain, and CaBP-9k expression is regulated by P4. We suggest that CaBP-9k expression regulated by steroid hormone may serve as an important regulator of cytosolic calcium concentration in the brain.
Collapse
Affiliation(s)
- Seon Young Park
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 362-763, Republic of Korea
| | - Yeong-Min Yoo
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 362-763, Republic of Korea
| | - Eui-Man Jung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 362-763, Republic of Korea.
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 362-763, Republic of Korea.
| |
Collapse
|
35
|
Sun G, Chen H, Liang WZ, Jan CR. Exploration of the effect of the alkaloid colchicine on Ca2+ handling and its related physiology in human oral cancer cells. Arch Oral Biol 2019; 102:179-185. [DOI: 10.1016/j.archoralbio.2019.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/15/2019] [Accepted: 04/27/2019] [Indexed: 10/26/2022]
|
36
|
Trzesniewski J, Altmann S, Jäger L, Kapfhammer JP. Reduced Purkinje cell size is compatible with near normal morphology and function of the cerebellar cortex in a mouse model of spinocerebellar ataxia. Exp Neurol 2018; 311:205-212. [PMID: 30312605 DOI: 10.1016/j.expneurol.2018.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/27/2018] [Accepted: 10/09/2018] [Indexed: 12/20/2022]
Abstract
Spinocerebellar ataxia type 14 (SCA14) is a dominantly inherited neurodegenerative disease caused by diverse mutations in the Protein Kinase C gamma (PKCγ) gene which is one of the crucial signaling molecules of Purkinje cells. We have previously created a mouse model of SCA14 by transgenic expression of a mutated PKCγ gene causing SCA14 with a mutation in the catalytic domain. Purkinje cells from the mutated mice have a strong reduction of their dendritic tree in organotypic slice cultures typical for increased PKC activity. There was no overt degeneration of Purkinje cells in vivo and the cerebellum appeared morphologically normal with the exception of lobule 7 where abnormal Purkinje cells were present. Besides from mild motor deficits the mice have no major phenotype. We have now done a more extensive study of cerebellar morphology in these mice and show by rapid Golgi staining that there is a marked reduction of Purkinje cell dendritic tree size throughout the cerebellum. Despite this reduction in dendritic tree size, climbing fiber innervation of Purkinje cells as visualized by immunostaining for the vesicular glutamate transporter 2 (vGlut2) appeared normal in most parts of the cerebellum. The same was true for the expression of the activity and plasticity markers pS6, c-Fos and Arc. These finding suggest that the cerebellar cortex in the transgenic mice is functioning fairly normal and that the reduction of dendritic tree size and the increased PKC activity can be compensated in most Purkinje cells. Around cerebellar lobule 7 there was high transgene expression from the L7 promotor and Purkinje cells showed abnormal morphologies. Climbing fiber innervation as well as the expression of the activity and plasticity markers was strongly disturbed in this area. Our results show that there is substantial potential for functional compensation in the cerebellar cortex. In lobule 7, an area with high transgene expression, compensation failed resulting in Purkinje cell degeneration and dysfunction.
Collapse
Affiliation(s)
- Jakub Trzesniewski
- Anatomical Institute, Department of Biomedicine Basel, University of Basel, Pestalozzistrasse 20, CH - 4056 Basel, Switzerland
| | - Sandrine Altmann
- Anatomical Institute, Department of Biomedicine Basel, University of Basel, Pestalozzistrasse 20, CH - 4056 Basel, Switzerland
| | - Levy Jäger
- Anatomical Institute, Department of Biomedicine Basel, University of Basel, Pestalozzistrasse 20, CH - 4056 Basel, Switzerland
| | - Josef P Kapfhammer
- Anatomical Institute, Department of Biomedicine Basel, University of Basel, Pestalozzistrasse 20, CH - 4056 Basel, Switzerland.
| |
Collapse
|
37
|
Orem BC, Pelisch N, Williams J, Nally JM, Stirling DP. Intracellular calcium release through IP 3 R or RyR contributes to secondary axonal degeneration. Neurobiol Dis 2017; 106:235-243. [DOI: 10.1016/j.nbd.2017.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/28/2017] [Accepted: 07/10/2017] [Indexed: 11/27/2022] Open
|