1
|
Sujkowski A, Ranxhi B, Bangash ZR, Chbihi ZM, Prifti MV, Qadri Z, Alam N, Todi SV, Tsou WL. Progressive degeneration in a new Drosophila model of spinocerebellar ataxia type 7. Sci Rep 2024; 14:14332. [PMID: 38906973 PMCID: PMC11192756 DOI: 10.1038/s41598-024-65172-4] [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: 11/10/2023] [Accepted: 06/18/2024] [Indexed: 06/23/2024] Open
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a progressive neurodegenerative disorder resulting from abnormal expansion of an uninterrupted polyglutamine (polyQ) repeat in its disease protein, ataxin-7 (ATXN7). ATXN7 is part of Spt-Ada-Gcn5 acetyltransferase (SAGA), an evolutionarily conserved transcriptional coactivation complex with critical roles in chromatin remodeling, cell signaling, neurodifferentiation, mitochondrial health and autophagy. SCA7 is dominantly inherited and characterized by genetic anticipation and high repeat-length instability. Patients with SCA7 experience progressive ataxia, atrophy, spasticity, and blindness. There is currently no cure for SCA7, and therapies are aimed at alleviating symptoms to increase quality of life. Here, we report novel Drosophila lines of SCA7 with polyQ repeats in wild-type and human disease patient range. We find that ATXN7 expression has age- and polyQ repeat length-dependent reduction in fruit fly survival and retinal instability, concomitant with increased ATXN7 protein aggregation. These new lines will provide important insight on disease progression that can be used in the future to identify therapeutic targets for SCA7 patients.
Collapse
Affiliation(s)
- Alyson Sujkowski
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Bedri Ranxhi
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Zoya R Bangash
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Zachary M Chbihi
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Matthew V Prifti
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Zaina Qadri
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Nadir Alam
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA.
| |
Collapse
|
2
|
Wang D, Honda S, Shin MK, Watase K, Mizusawa H, Ishikawa K, Shimizu S. Subcellular localization and ER-mediated cytotoxic function of α1A and α1ACT in spinocerebellar ataxia type 6. Biochem Biophys Res Commun 2024; 695:149481. [PMID: 38211534 DOI: 10.1016/j.bbrc.2024.149481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Spinocerebellar ataxia type 6 (SCA6) is a polyglutamine (polyQ) disease, which is caused by the elongation of CAG repeats encoding polyQ in the CACNA1A gene. The CACNA1A gene encodes two proteins, namely, α1A (a subunit of the plasma membrane calcium channel), which is translated in its entire length, and α1ACT, which is translated from the second cistron, and both proteins have a polyQ tract. The α1A-polyQ and α1ACT-polyQ proteins with an elongated polyQ stretch have been reported to form aggregates in cells and induce neuronal cell death, but the subcellular localization of these proteins and their cytotoxic properties remain unclear. In this study, we first analyzed SCA6 model mice and found that α1A-polyQlong localized mainly to the Golgi apparatus, whereas a portion of α1ACT-polyQlong localized to the nucleus. Analysis using Neuro2a cells also showed similar subcellular localizations of these proteins, and a proportion of both proteins localized to the endoplasmic reticulum (ER). Cytotoxic studies demonstrated that both proteins induce both the ER stress response and apoptosis, indicating that they are able to induce ER stress-induced apoptosis.
Collapse
Affiliation(s)
- Di Wang
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Department of Personalized Genomic Medicine for Health, Graduate School of Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Shinya Honda
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Min Kyoung Shin
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kei Watase
- Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Hidehiro Mizusawa
- Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kinya Ishikawa
- Department of Personalized Genomic Medicine for Health, Graduate School of Medicine, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Shigeomi Shimizu
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
3
|
Kumar M, Tyagi N, Faruq M. The molecular mechanisms of spinocerebellar ataxias for DNA repeat expansion in disease. Emerg Top Life Sci 2023; 7:289-312. [PMID: 37668011 DOI: 10.1042/etls20230013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023]
Abstract
Spinocerebellar ataxias (SCAs) are a heterogenous group of neurodegenerative disorders which commonly inherited in an autosomal dominant manner. They cause muscle incoordination due to degeneration of the cerebellum and other parts of nervous system. Out of all the characterized (>50) SCAs, 14 SCAs are caused due to microsatellite repeat expansion mutations. Repeat expansions can result in toxic protein gain-of-function, protein loss-of-function, and/or RNA gain-of-function effects. The location and the nature of mutation modulate the underlying disease pathophysiology resulting in varying disease manifestations. Potential toxic effects of these mutations likely affect key major cellular processes such as transcriptional regulation, mitochondrial functioning, ion channel dysfunction and synaptic transmission. Involvement of several common pathways suggests interlinked function of genes implicated in the disease pathogenesis. A better understanding of the shared and distinct molecular pathogenic mechanisms in these diseases is required to develop targeted therapeutic tools and interventions for disease management. The prime focus of this review is to elaborate on how expanded 'CAG' repeats contribute to the common modes of neurotoxicity and their possible therapeutic targets in management of such devastating disorders.
Collapse
Affiliation(s)
- Manish Kumar
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| | - Nishu Tyagi
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| | - Mohammed Faruq
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| |
Collapse
|
4
|
Sujkowski AL, Ranxhi B, Prifti MV, Alam N, Todi SV, Tsou WL. Progressive degeneration in a new Drosophila model of Spinocerebellar Ataxia type 7. RESEARCH SQUARE 2023:rs.3.rs-3592641. [PMID: 38045332 PMCID: PMC10690306 DOI: 10.21203/rs.3.rs-3592641/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a progressive neurodegenerative disorder resulting from abnormal expansion of polyglutamine (polyQ) in its disease protein, ataxin-7 (ATXN7). ATXN7 is part of Spt-Ada-Gcn5 acetyltransferase (SAGA), an evolutionarily conserved transcriptional coactivation complex with critical roles in chromatin remodeling, cell signaling, neurodifferentiation, mitochondrial health and autophagy. SCA7 is dominantly inherited and characterized by genetic anticipation and high repeat-length instability. Patients with SCA7 experience progressive ataxia, atrophy, spasticity, and blindness. There is currently no cure for SCA7, and therapies are aimed at alleviating symptoms to increase quality of life. Here, we report novel Drosophila lines of SCA7 with polyQ repeats in wild-type and human disease patient range. We find that ATXN7 expression has age- and polyQ repeat length-dependent reduction in survival and retinal instability, concomitant with increased ATXN7 protein aggregation. These new lines will provide important insight on disease progression that can be used in the future to identify therapeutic targets for SCA7 patients.
Collapse
Affiliation(s)
| | - Bedri Ranxhi
- Department of Pharmacology, Wayne State University School of Medicine
| | - Matthew V Prifti
- Department of Pharmacology, Wayne State University School of Medicine
| | - Nadir Alam
- Department of Pharmacology, Wayne State University School of Medicine
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University School of Medicine
- Department of Neurology, Wayne State University School of Medicine
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University School of Medicine
| |
Collapse
|
5
|
Sujkowski AL, Ranxhi B, Prifti MV, Alam N, Todi SV, Tsou WL. Progressive degeneration in a new Drosophila model of Spinocerebellar Ataxia type 7. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566106. [PMID: 37986914 PMCID: PMC10659390 DOI: 10.1101/2023.11.07.566106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a progressive neurodegenerative disorder resulting from abnormal expansion of polyglutamine (polyQ) in its disease protein, ataxin-7 (ATXN7). ATXN7 is part of Spt-Ada-Gcn5 acetyltransferase (SAGA), an evolutionarily conserved transcriptional coactivation complex with critical roles in chromatin remodeling, cell signaling, neurodifferentiation, mitochondrial health and autophagy. SCA7 is dominantly inherited and characterized by genetic anticipation and high repeat-length instability. Patients with SCA7 experience progressive ataxia, atrophy, spasticity, and blindness. There is currently no cure for SCA7, and therapies are aimed at alleviating symptoms to increase quality of life. Here, we report novel Drosophila lines of SCA7 with polyQ repeats in wild-type and human disease patient range. We find that ATXN7 expression has age- and polyQ repeat length-dependent reduction in survival and retinal instability, concomitant with increased ATXN7 protein aggregation. These new lines will provide important insight on disease progression that can be used in the future to identify therapeutic targets for SCA7 patients.
Collapse
|
6
|
Kramer AA, Bennett DF, Barañano KW, Bannister RA. A neurodevelopmental disorder caused by a dysfunctional CACNA1A allele. eNeurologicalSci 2023; 31:100456. [PMID: 36938367 PMCID: PMC10020665 DOI: 10.1016/j.ensci.2023.100456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/05/2022] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
P/Q-type Ca2+ flux into nerve terminals via CaV2.1 channels is essential for neurotransmitter release at neuromuscular junctions and nearly all central synapses. Mutations in CACNA1A, the gene encoding CaV2.1, cause a spectrum of pediatric neurological disorders. We have identified a patient harboring an autosomal-dominant de novo frameshift-causing nucleotide duplication in CACNA1A (c.5018dupG). The duplicated guanine precipitated 43 residues of altered amino acid sequence beginning with a glutamine to serine substitution in CaV2.1 at position 1674 ending with a premature stop codon (CaV2.1 p.Gln1674Serfs*43). The patient presented with episodic downbeat vertical nystagmus, hypotonia, ataxia, developmental delay and febrile seizures. In patch-clamp experiments, no Ba2+ current was observed in tsA-201 cells expressing CaV2.1 p.Gln1674Serfs*43 with β4 and α2δ-1 auxiliary subunits. The ablation of divalent flux in response to depolarization was likely attributable to the inability of CaV2.1 p.Gln1674Serfs*43 to form a complete channel pore. Our results suggest that the pathology resulting from this frameshift-inducing nucleotide duplication is a consequence of an effective haploinsufficiency.
Collapse
Affiliation(s)
- Audra A. Kramer
- Department of Pathology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA
- Corresponding authors at: National Institutes of Health, Center for Scientific Review, Division of Neuroscience, Development and Aging, 6701 Rockledge Drive, Bethesda, MD 20892, USA.
| | - Daniel F. Bennett
- Department of Pathology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA
| | - Kristin W. Barañano
- Department of Neurology, Johns Hopkins University School of Medicine, 200 North Wolfe Street, Suite 2158, Baltimore, MD 21287, USA
| | - Roger A. Bannister
- Department of Pathology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA
- Corresponding authors at: National Institutes of Health, Center for Scientific Review, Division of Neuroscience, Development and Aging, 6701 Rockledge Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
7
|
Bohne P, Rybarski M, Mourabit DBE, Krause F, Mark MD. Cerebellar contribution to threat probability in a SCA6 mouse model. Hum Mol Genet 2022; 31:3807-3828. [PMID: 35708512 PMCID: PMC9652111 DOI: 10.1093/hmg/ddac135] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/24/2022] [Accepted: 06/09/2022] [Indexed: 02/05/2023] Open
Abstract
Fear and anxiety have proven to be essential during the evolutionary process. However, the mechanisms involved in recognizing and categorizing threat probability (i.e. low to high) to elicit the appropriate defensive behavior are yet to be determined. In this study, we investigated the cerebellar contribution in evoking appropriate defensive escape behavior using a purely cerebellar, neurodegenerative mouse model for spinocerebellar ataxia type 6 which is caused by an expanded CAG repeat in exon 47 of the P/Q type calcium channel α1A subunit. These mice overexpress the carboxy terminus (CT) of the P/Q type calcium channel containing an expanded 27 CAG repeat specifically in cerebellar Purkinje cells (CT-longQ27PC). We found that our CT-longQ27PC mice exhibit anxiolytic behavior in the open field, elevated plus maze and light/dark place preference tests, which could be recovered with more threatening conditions such as brighter lighting, meowing sounds and an ultrasound repellent. Their innate fear to find safety in the Barnes maze and visual cliff tests was also diminished with subsequent trials, which could be partially recovered with an ultrasound repellent in the Barnes maze. However, under higher threat conditions such as in the light/dark place preference with ultrasound repellent and in the looming tests, CT-longQ27PC mice responded with higher defensive escape behaviors as controls. Moreover, CT-longQ27PC mice displayed increased levels of CT-labeled aggregates compared with controls. Together these data suggest that cerebellar degeneration by overexpression of CT-longQ27PC is sufficient to impair defensive escape responses in those mice.
Collapse
Affiliation(s)
- Pauline Bohne
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum D-44780, Germany
| | - Max Rybarski
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum D-44780, Germany
| | | | - Felix Krause
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum D-44780, Germany
| | - Melanie D Mark
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum D-44780, Germany
| |
Collapse
|
8
|
Incebacak Eltemur RD, Nguyen HP, Weber JJ. Calpain-mediated proteolysis as driver and modulator of polyglutamine toxicity. Front Mol Neurosci 2022; 15:1020104. [PMID: 36385755 PMCID: PMC9648470 DOI: 10.3389/fnmol.2022.1020104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 09/22/2023] Open
Abstract
Among posttranslational modifications, directed proteolytic processes have the strongest impact on protein integrity. They are executed by a variety of cellular machineries and lead to a wide range of molecular consequences. Compared to other forms of proteolytic enzymes, the class of calcium-activated calpains is considered as modulator proteases due to their limited proteolytic activity, which changes the structure and function of their target substrates. In the context of neurodegeneration and - in particular - polyglutamine disorders, proteolytic events have been linked to modulatory effects on the molecular pathogenesis by generating harmful breakdown products of disease proteins. These findings led to the formulation of the toxic fragment hypothesis, and calpains appeared to be one of the key players and auspicious therapeutic targets in Huntington disease and Machado Joseph disease. This review provides a current survey of the role of calpains in proteolytic processes found in polyglutamine disorders. Together with insights into general concepts behind toxic fragments and findings in polyglutamine disorders, this work aims to inspire researchers to broaden and deepen the knowledge in this field, which will help to evaluate calpain-mediated proteolysis as a unifying and therapeutically targetable posttranslational mechanism in neurodegeneration.
Collapse
Affiliation(s)
- Rana Dilara Incebacak Eltemur
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Jonasz Jeremiasz Weber
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Johnson SL, Tsou WL, Prifti MV, Harris AL, Todi SV. A survey of protein interactions and posttranslational modifications that influence the polyglutamine diseases. Front Mol Neurosci 2022; 15:974167. [PMID: 36187346 PMCID: PMC9515312 DOI: 10.3389/fnmol.2022.974167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/27/2022] [Indexed: 01/20/2023] Open
Abstract
The presence and aggregation of misfolded proteins has deleterious effects in the nervous system. Among the various diseases caused by misfolded proteins is the family of the polyglutamine (polyQ) disorders. This family comprises nine members, all stemming from the same mutation—the abnormal elongation of a polyQ repeat in nine different proteins—which causes protein misfolding and aggregation, cellular dysfunction and disease. While it is the same type of mutation that causes them, each disease is distinct: it is influenced by regions and domains that surround the polyQ repeat; by proteins with which they interact; and by posttranslational modifications they receive. Here, we overview the role of non-polyQ regions that control the pathogenicity of the expanded polyQ repeat. We begin by introducing each polyQ disease, the genes affected, and the symptoms experienced by patients. Subsequently, we provide a survey of protein-protein interactions and posttranslational modifications that regulate polyQ toxicity. We conclude by discussing shared processes and pathways that bring some of the polyQ diseases together and may serve as common therapeutic entry points for this family of incurable disorders.
Collapse
Affiliation(s)
- Sean L. Johnson
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Matthew V. Prifti
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Autumn L. Harris
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
- Maximizing Access to Research Careers (MARC) Program, Wayne State University, Detroit, MI, United States
| | - Sokol V. Todi
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
- Maximizing Access to Research Careers (MARC) Program, Wayne State University, Detroit, MI, United States
- Department of Neurology, Wayne State University, Detroit, MI, United States
- *Correspondence: Sokol V. Todi,
| |
Collapse
|
10
|
Heck J, Palmeira Do Amaral AC, Weißbach S, El Khallouqi A, Bikbaev A, Heine M. More than a pore: How voltage-gated calcium channels act on different levels of neuronal communication regulation. Channels (Austin) 2021; 15:322-338. [PMID: 34107849 PMCID: PMC8205089 DOI: 10.1080/19336950.2021.1900024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) represent key regulators of the calcium influx through the plasma membrane of excitable cells, like neurons. Activated by the depolarization of the membrane, the opening of VGCCs induces very transient and local changes in the intracellular calcium concentration, known as calcium nanodomains, that in turn trigger calcium-dependent signaling cascades and the release of chemical neurotransmitters. Based on their central importance as concierges of excitation-secretion coupling and therefore neuronal communication, VGCCs have been studied in multiple aspects of neuronal function and malfunction. However, studies on molecular interaction partners and recent progress in omics technologies have extended the actual concept of these molecules. With this review, we want to illustrate some new perspectives of VGCCs reaching beyond their function as calcium-permeable pores in the plasma membrane. Therefore, we will discuss the relevance of VGCCs as voltage sensors in functional complexes with ryanodine receptors, channel-independent actions of auxiliary VGCC subunits, and provide an insight into how VGCCs even directly participate in gene regulation. Furthermore, we will illustrate how structural changes in the intracellular C-terminus of VGCCs generated by alternative splicing events might not only affect the biophysical channel characteristics but rather determine their molecular environment and downstream signaling pathways.
Collapse
Affiliation(s)
- Jennifer Heck
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Ana Carolina Palmeira Do Amaral
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Stephan Weißbach
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
- Computational Genomics and Bioinformatics, Johannes Gutenberg-University Mainz, University Medical Center Mainz, Institute for Human Genetics, Mainz, Germany
| | - Abderazzaq El Khallouqi
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Arthur Bikbaev
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| | - Martin Heine
- Functional Neurobiology, Johannes Gutenberg-University Mainz, Institute for Developmental Biology and Neurobiology, Mainz, Germany
| |
Collapse
|
11
|
Donaldson J, Powell S, Rickards N, Holmans P, Jones L. What is the Pathogenic CAG Expansion Length in Huntington's Disease? J Huntingtons Dis 2021; 10:175-202. [PMID: 33579866 PMCID: PMC7990448 DOI: 10.3233/jhd-200445] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Huntington's disease (HD) (OMIM 143100) is caused by an expanded CAG repeat tract in the HTT gene. The inherited CAG length is known to expand further in somatic and germline cells in HD subjects. Age at onset of the disease is inversely correlated with the inherited CAG length, but is further modulated by a series of genetic modifiers which are most likely to act on the CAG repeat in HTT that permit it to further expand. Longer repeats are more prone to expansions, and this expansion is age dependent and tissue-specific. Given that the inherited tract expands through life and most subjects develop disease in mid-life, this implies that in cells that degenerate, the CAG length is likely to be longer than the inherited length. These findings suggest two thresholds- the inherited CAG length which permits further expansion, and the intracellular pathogenic threshold, above which cells become dysfunctional and die. This two-step mechanism has been previously proposed and modelled mathematically to give an intracellular pathogenic threshold at a tract length of 115 CAG (95% confidence intervals 70- 165 CAG). Empirically, the intracellular pathogenic threshold is difficult to determine. Clues from studies of people and models of HD, and from other diseases caused by expanded repeat tracts, place this threshold between 60- 100 CAG, most likely towards the upper part of that range. We assess this evidence and discuss how the intracellular pathogenic threshold in manifest disease might be better determined. Knowing the cellular pathogenic threshold would be informative for both understanding the mechanism in HD and deploying treatments.
Collapse
Affiliation(s)
- Jasmine Donaldson
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Sophie Powell
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Nadia Rickards
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Peter Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Lesley Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
12
|
Yeow SQZ, Loh KWZ, Soong TW. Calcium Channel Splice Variants and Their Effects in Brain and Cardiovascular Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:67-86. [DOI: 10.1007/978-981-16-4254-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
13
|
Ménard C, Charnet P, Rousset M, Vignes M, Cens T. Cav2.1 C-terminal fragments produced in Xenopus laevis oocytes do not modify the channel expression and functional properties. Eur J Neurosci 2020; 51:1900-1913. [PMID: 31981388 DOI: 10.1111/ejn.14685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 12/01/2022]
Abstract
The sequence and genomic organization of the CACNA1A gene that encodes the Cav2.1 subunit of both P and Q-type Ca2+ channels are well conserved in mammals. In human, rat and mouse CACNA1A, the use of an alternative acceptor site at the exon 46-47 boundary results in the expression of a long Cav2.1 splice variant. In transfected cells, the long isoform of human Cav2.1 produces a C-terminal fragment, but it is not known whether this fragment affects Cav2.1 expression or functional properties. Here, we cloned the long isoform of rat Cav2.1 (Cav2.1(e47)) and identified a novel variant with a shorter C-terminus (Cav2.1(e47s)) that differs from those previously described in the rat and mouse. When expressed in Xenopus laevis oocytes, Cav2.1(e47) and Cav2.1(e47s) displayed similar functional properties as the short isoform (Cav2.1). We show that Cav2.1 isoforms produced short (CT1) and long (CT1(e47)) C-terminal fragments that interacted in vivo with the auxiliary Cavβ4a subunit. Overexpression of the C-terminal fragments did not affect Cav2.1 expression and functional properties. Furthermore, the functional properties of a Cav2.1 mutant without the C-terminal Cavβ4 binding domain (Cav2.1ΔCT2) were similar to those of Cav2.1 and were not influenced by the co-expression of the missing fragments (CT2 or CT2(e47)). Our results exclude a functional role of the C-terminal fragments in Cav2.1 biophysical properties in an expression system widely used to study this channel.
Collapse
Affiliation(s)
- Claudine Ménard
- Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,IBMM, Université de Montpellier, Montpellier, France
| | - Pierre Charnet
- Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,IBMM, Université de Montpellier, Montpellier, France
| | - Matthieu Rousset
- Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,IBMM, Université de Montpellier, Montpellier, France
| | - Michel Vignes
- Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,IBMM, Université de Montpellier, Montpellier, France
| | - Thierry Cens
- Institut des Biomolécules Max Mousseron (IBMM), Montpellier, France.,IBMM, Université de Montpellier, Montpellier, France
| |
Collapse
|
14
|
Verma AK, Khan E, Bhagwat SR, Kumar A. Exploring the Potential of Small Molecule-Based Therapeutic Approaches for Targeting Trinucleotide Repeat Disorders. Mol Neurobiol 2019; 57:566-584. [DOI: 10.1007/s12035-019-01724-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022]
|
15
|
Aikawa T, Watanabe T, Miyazaki T, Mikuni T, Wakamori M, Sakurai M, Aizawa H, Ishizu N, Watanabe M, Kano M, Mizusawa H, Watase K. Alternative splicing in the C-terminal tail of Cav2.1 is essential for preventing a neurological disease in mice. Hum Mol Genet 2018; 26:3094-3104. [PMID: 28510727 DOI: 10.1093/hmg/ddx193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 05/09/2017] [Indexed: 12/13/2022] Open
Abstract
Alternative splicing (AS) that occurs at the final coding exon (exon 47) of the Cav2.1 voltage-gated calcium channel (VGCC) gene produces two major isoforms in the brain, MPI and MPc. These isoforms differ in their splice acceptor sites; human MPI is translated into a polyglutamine tract associated with spinocerebellar ataxia type 6 (SCA6), whereas MPc splices to an immediate stop codon, resulting in a shorter cytoplasmic tail. To gain insight into the functional role of the AS in vivo and whether modulating the splice patterns at this locus can be a potential therapeutic strategy for SCA6, here we created knockin mice that exclusively express MPc by inserting the splice-site mutation. The resultant Cacna1aCtmKO/CtmKO mice developed non-progressive neurological phenotypes, featuring early-onset ataxia and absence seizure without significant alterations in the basic properties of the channel. Interactions of Cav2.1 with Cavβ4 and Rimbp2 were significantly reduced while those with GABAB2 were enhanced in the cerebellum of Cacna1aCtmKO/CtmKO mice. Treatment with the GABAB antagonist CGP35348 partially rescued the motor impairments seen in Cacna1aCtmKO/CtmKO mice. These results suggest that the carboxyl-terminal domain of Cav2.1 is not essential for maintaining the basic properties of the channel in the cerebellar Purkinje neurons but is involved in multiple interactions of Cav2.1 with other proteins, and plays an essential role in preventing a complex neurological disease.
Collapse
Affiliation(s)
- Tomonori Aikawa
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan
| | - Takaki Watanabe
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Taisuke Miyazaki
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | - Takayasu Mikuni
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.,Max Planck Florida Institute for Neuroscience, Max Planck Way Jupiter, FL 33458, USA
| | - Minoru Wakamori
- Department of Oral Biology, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan
| | - Miyano Sakurai
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Hidenori Aizawa
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Nobutaka Ishizu
- Department of Neurology, Tokyo National Hospital, Tokyo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Hidehiro Mizusawa
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan.,Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
| | - Kei Watase
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan
| |
Collapse
|
16
|
Miyazaki Y, Du X, Muramatsu SI, Gomez CM. An miRNA-mediated therapy for SCA6 blocks IRES-driven translation of the CACNA1A second cistron. Sci Transl Med 2017; 8:347ra94. [PMID: 27412786 DOI: 10.1126/scitranslmed.aaf5660] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/21/2016] [Indexed: 12/17/2022]
Abstract
Spinocerebellar ataxia type 6 (SCA6) is a dominantly inherited neurodegenerative disease characterized by slowly progressive ataxia and Purkinje cell degeneration. SCA6 is caused by a polyglutamine repeat expansion within a second CACNA1A gene product, α1ACT. α1ACT expression is under the control of an internal ribosomal entry site (IRES) present within the CACNA1A coding region. Whereas SCA6 allele knock-in mice show indistinguishable phenotypes from wild-type littermates, expression of SCA6-associated α1ACT (α1ACTSCA6) driven by a Purkinje cell-specific promoter in mice produces slowly progressive ataxia and cerebellar atrophy. We developed an early-onset SCA6 mouse model using an adeno-associated virus (AAV)-based gene delivery system to ectopically express CACNA1A IRES-driven α1ACTSCA6 to test the potential of CACNA1A IRES-targeting therapies. Mice expressing AAV9-mediated CACNA1A IRES-driven α1ACTSCA6 exhibited early-onset ataxia, motor deficits, and Purkinje cell degeneration. We identified miR-3191-5p as a microRNA (miRNA) that targeted CACNA1A IRES and preferentially inhibited the CACNA1A IRES-driven translation of α1ACT in an Argonaute 4 (Ago4)-dependent manner. We found that eukaryotic initiation factors (eIFs), eIF4AII and eIF4GII, interacted with the CACNA1A IRES to enhance α1ACT translation. Ago4-bound miR-3191-5p blocked the interaction of eIF4AII and eIF4GII with the CACNA1A IRES, attenuating IRES-driven α1ACT translation. Furthermore, AAV9-mediated delivery of miR-3191-5p protected mice from the ataxia, motor deficits, and Purkinje cell degeneration caused by CACNA1A IRES-driven α1ACTSCA6 We have established proof of principle that viral delivery of an miRNA can rescue a disease phenotype through modulation of cellular IRES activity in a mouse model.
Collapse
Affiliation(s)
- Yu Miyazaki
- Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Xiaofei Du
- Department of Neurology, University of Chicago, Chicago, IL 60637, USA
| | - Shin-Ichi Muramatsu
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi 3290498, Japan. Center for Gene and Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo 1088639, Japan
| | | |
Collapse
|
17
|
Bavassano C, Eigentler A, Stanika R, Obermair GJ, Boesch S, Dechant G, Nat R. Bicistronic CACNA1A Gene Expression in Neurons Derived from Spinocerebellar Ataxia Type 6 Patient-Induced Pluripotent Stem Cells. Stem Cells Dev 2017; 26:1612-1625. [PMID: 28946818 PMCID: PMC5684673 DOI: 10.1089/scd.2017.0085] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Spinocerebellar ataxia type 6 (SCA6) is an autosomal-dominant neurodegenerative disorder that is caused by a CAG trinucleotide repeat expansion in the CACNA1A gene. As one of the few bicistronic genes discovered in the human genome, CACNA1A encodes not only the α1A subunit of the P/Q type voltage-gated Ca2+ channel CaV2.1 but also the α1ACT protein, a 75 kDa transcription factor sharing the sequence of the cytoplasmic C-terminal tail of the α1A subunit. Isoforms of both proteins contain the polyglutamine (polyQ) domain that is expanded in SCA6 patients. Although certain SCA6 phenotypes appear to be specific for Purkinje neurons, other pathogenic effects of the SCA6 polyQ mutation can affect a broad spectrum of central nervous system (CNS) neuronal subtypes. We investigated the expression and function of CACNA1A gene products in human neurons derived from induced pluripotent stem cells from two SCA6 patients. Expression levels of CACNA1A encoding α1A subunit were similar between SCA6 and control neurons, and no differences were found in the subcellular distribution of CaV2.1 channel protein. The α1ACT immunoreactivity was detected in the majority of cell nuclei of SCA6 and control neurons. Although no SCA6 genotype-dependent differences in CaV2.1 channel function were observed, they were found in the expression levels of the α1ACT target gene Granulin (GRN) and in glutamate-induced cell vulnerability.
Collapse
Affiliation(s)
- Carlo Bavassano
- 1 Institute for Neuroscience, Medical University of Innsbruck , Innsbruck, Austria
| | - Andreas Eigentler
- 1 Institute for Neuroscience, Medical University of Innsbruck , Innsbruck, Austria
| | - Ruslan Stanika
- 2 Division of Physiology, Medical University of Innsbruck , Innsbruck, Austria
| | - Gerald J Obermair
- 2 Division of Physiology, Medical University of Innsbruck , Innsbruck, Austria
| | - Sylvia Boesch
- 3 Department of Neurology, Medical University of Innsbruck , Innsbruck, Austria
| | - Georg Dechant
- 1 Institute for Neuroscience, Medical University of Innsbruck , Innsbruck, Austria
| | - Roxana Nat
- 1 Institute for Neuroscience, Medical University of Innsbruck , Innsbruck, Austria
| |
Collapse
|
18
|
Nazıroğlu M, Blum W, Jósvay K, Çiğ B, Henzi T, Oláh Z, Vizler C, Schwaller B, Pecze L. Menthol evokes Ca 2+ signals and induces oxidative stress independently of the presence of TRPM8 (menthol) receptor in cancer cells. Redox Biol 2017; 14:439-449. [PMID: 29078169 PMCID: PMC5680524 DOI: 10.1016/j.redox.2017.10.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/11/2017] [Indexed: 12/27/2022] Open
Abstract
Menthol is a naturally occurring monoterpene alcohol possessing remarkable biological properties including antipruritic, analgesic, antiseptic, anti-inflammatory and cooling effects. Here, we examined the menthol-evoked Ca2+ signals in breast and prostate cancer cell lines. The effect of menthol (50–500 µM) was predicted to be mediated by the transient receptor potential ion channel melastatin subtype 8 (TRPM8). However, the intensity of menthol-evoked Ca2+ signals did not correlate with the expression levels of TRPM8 in breast and prostate cancer cells indicating a TRPM8-independent signaling pathway. Menthol-evoked Ca2+ signals were analyzed in detail in Du 145 prostate cancer cells, as well as in CRISPR/Cas9 TRPM8-knockout Du 145 cells. Menthol (500 µM) induced Ca2+ oscillations in both cell lines, thus independent of TRPM8, which were however dependent on the production of inositol trisphosphate. Results based on pharmacological tools point to an involvement of the purinergic pathway in menthol-evoked Ca2+ responses. Finally, menthol (50–500 µM) decreased cell viability and induced oxidative stress independently of the presence of TRPM8 channels, despite that temperature-evoked TRPM8-mediated inward currents were significantly decreased in TRPM8-knockout Du 145 cells compared to wild type Du 145 cells.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey; Department of Biophysics, Faculty of Mediciene, Suleyman Demirel University, Isparta, Turkey
| | - Walter Blum
- Anatomy, Department of Medicine, University of Fribourg, Route Albert-Gockel 1, Fribourg, Switzerland
| | - Katalin Jósvay
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Bilal Çiğ
- Department of Biophysics, Faculty of Mediciene, Suleyman Demirel University, Isparta, Turkey
| | - Thomas Henzi
- Anatomy, Department of Medicine, University of Fribourg, Route Albert-Gockel 1, Fribourg, Switzerland
| | - Zoltán Oláh
- Institute of Chemistry, Faculty of Materials Science and Engineering, University of Miskolc, Miskolc-Egyetemváros, Hungary; Acheuron Ltd., Szeged, Hungary
| | - Csaba Vizler
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Beat Schwaller
- Anatomy, Department of Medicine, University of Fribourg, Route Albert-Gockel 1, Fribourg, Switzerland
| | - László Pecze
- Anatomy, Department of Medicine, University of Fribourg, Route Albert-Gockel 1, Fribourg, Switzerland.
| |
Collapse
|
19
|
Soga K, Ishikawa K, Furuya T, Iida T, Yamada T, Ando N, Ota K, Kanno-Okada H, Tanaka S, Shintaku M, Eishi Y, Mizusawa H, Yokota T. Gene dosage effect in spinocerebellar ataxia type 6 homozygotes: A clinical and neuropathological study. J Neurol Sci 2016; 373:321-328. [PMID: 28131213 DOI: 10.1016/j.jns.2016.12.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/20/2016] [Accepted: 12/23/2016] [Indexed: 02/07/2023]
Abstract
Spinocerebellar ataxia type 6 (SCA6) is an autosomal dominant neurodegenerative disorder. However, it remains unclear whether SCA6 shows a gene dosage effect, defined by earlier age-of-onset in homozygotes than heterozygotes. Herein, we retrospectively analyzed four homozygous SCA6 subjects from our single institution cohort of 120 SCA6 subjects. We also performed a neuropathological investigation into an SCA6 individual with compound heterozygous expansions. In the 116 heterozygotes, there was an inverse correlation of age-of-onset with the number of CAG repeats in the expanded allele, and with the total number of CAG repeats, in both normal and expanded alleles. The age-of-onset in the four homozygotes was within the 95% confidence interval of the age-of-onset versus the repeat-lengths correlations determined in the 116 heterozygotes. Nevertheless, all homozygotes had earlier onset than their parents, and showed rapid disease progression. Neuropathology revealed neuronal loss, as well as α1A-calcium channel protein aggregates in Purkinje cells, a few α1A-calcium channel protein aggregates in the neocortex and basal ganglia, and neuronal loss in Clarke's column and the globus pallidus not seen in heterozygotes. These data suggest a mild clinical and neuropathological gene dosage effect in SCA6 subjects.
Collapse
Affiliation(s)
- Kazumasa Soga
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Kinya Ishikawa
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan; The Center for Personalized Medicine for Healthy Aging, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan.
| | - Tokuro Furuya
- Department of Neurology, Kawaguchi Kogyo General Hospital, 1-18-15 Aoki, Kawaguchi, Saitama 332-0031, Japan
| | - Tadatsune Iida
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan; Department of Cellular Neurobiology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tetsuo Yamada
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan; Laboratory of Pathology, Department of Clinical Laboratory Medicine, Bunkyo Gakuin University Graduate School of Health Care Science, 2-4-1 Mukogaoka, Bunkyo-ku, Tokyo 113-0023, Japan
| | - Noboru Ando
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Kiyobumi Ota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Hiromi Kanno-Okada
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, North 15, West 7, Kita-ku, Sapporo 060-8638, Hokkaido, Japan; Department of Surgical Pathology, Hokkaido University Hospital, Hokkaido University, North 14, West 5, Kita-ku, Sapporo 060-8648, Hokkaido, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, North 15, West 7, Kita-ku, Sapporo 060-8638, Hokkaido, Japan
| | - Masayuki Shintaku
- Department of Pathology, Shiga Medical Center for Adults, 5-4-30 Moriyama, Moriyama, Shiga 524-8524, Japan
| | - Yoshinobu Eishi
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Hidehiro Mizusawa
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan; The National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8551, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| |
Collapse
|
20
|
Modulation of Molecular Chaperones in Huntington’s Disease and Other Polyglutamine Disorders. Mol Neurobiol 2016; 54:5829-5854. [DOI: 10.1007/s12035-016-0120-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/12/2016] [Indexed: 12/20/2022]
|
21
|
Endogenous TRPV1 stimulation leads to the activation of the inositol phospholipid pathway necessary for sustained Ca 2+ oscillations. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2905-2915. [PMID: 27663071 DOI: 10.1016/j.bbamcr.2016.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/05/2016] [Accepted: 09/16/2016] [Indexed: 01/15/2023]
Abstract
Sensory neuron subpopulations as well as breast and prostate cancer cells express functional transient receptor potential vanilloid type 1 (TRPV1) ion channels; however little is known how TRPV1 activation leads to biological responses. Agonist-induced activation of TRPV1 resulted in specific spatiotemporal patterns of cytoplasmic Ca2+ signals in breast and prostate cancer-derived cells. Capsaicin (CAPS; 50μM) evoked intracellular Ca2+ oscillations and/or intercellular Ca2+ waves in all cell lines. As evidenced in prostate cancer Du 145 cells, oscillations were largely dependent on the expression of functional TRPV1 channels in the plasma membrane, phospholipase C activation and on the presence of extracellular Ca2+ ions. Concomitant oscillations of the mitochondrial matrix Ca2+ concentration resulted in mitochondria energization evidenced by increased ATP production. CAPS-induced Ca2+ oscillations also occurred in a subset of sensory neurons, yet already at lower CAPS concentrations (1μM). Stimulation of ectopically expressed TRPV1 channels in CAPS-insensitive NIH-3T3 cells didn't provoke CAPS-triggered Ca2+ oscillations; rather it resulted in low-magnitude, long-lasting elevations of the cytosolic Ca2+ concentration. This indicates that sole TRPV1 activation is not sufficient to generate Ca2+ oscillations. Instead the initial TRPV1-mediated signal leads to the activation of the inositol phospholipid pathway. This in turn suffices to generate a biologically relevant frequency-modulated Ca2+ signal.
Collapse
|
22
|
Mark MD, Schwitalla JC, Groemmke M, Herlitze S. Keeping Our Calcium in Balance to Maintain Our Balance. Biochem Biophys Res Commun 2016; 483:1040-1050. [PMID: 27392710 DOI: 10.1016/j.bbrc.2016.07.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/04/2016] [Indexed: 01/13/2023]
Abstract
Calcium is a key signaling molecule and ion involved in a variety of diverse processes in our central nervous system (CNS) which include gene expression, synaptic transmission and plasticity, neuronal excitability and cell maintenance. Proper control of calcium signaling is not only vital for neuronal physiology but also cell survival. Mutations in fundamental channels, transporters and second messenger proteins involved in orchestrating the balance of our calcium homeostasis can lead to severe neurodegenerative disorders, such as Spinocerebellar (SCA) and Episodic (EA) ataxias. Hereditary ataxias make up a remarkably diverse group of neurological disorders clinically characterized by gait ataxia, nystagmus, dysarthria, trunk and limb ataxia and often atrophy of the cerebellum. The largest family of hereditary ataxias is SCAs which consists of a growing family of 42 members. A relatively smaller family of 8 members compose the EAs. The gene mutations responsible for half of the EA members and over 35 of the SCA subtypes have been identified, and several have been found to be responsible for cerebellar atrophy, abnormal intracellular calcium levels, dysregulation of Purkinje cell pacemaking, altered cerebellar synaptic transmission and/or ataxia in mouse models. Although the genetic diversity and affected cellular pathways of hereditary ataxias are broad, one common theme amongst these genes is their effects on maintaining calcium balance in primarily the cerebellum. There is emerging evidence that the pathogenesis of hereditary ataxias may be caused by imbalances in intracellular calcium due to genetic mutations in calcium-mediating proteins. In this review we will discuss the current evidence supporting the role of deranged calcium as the culprit to neurodegenerative diseases with a primary focus on SCAs and EAs.
Collapse
Affiliation(s)
- Melanie D Mark
- Department of Zoology and Neurobiology, ND7/31, Ruhr University Bochum, Universitätsstr. 150, D-44780 Bochum, Germany.
| | - Jan Claudius Schwitalla
- Department of Zoology and Neurobiology, ND7/31, Ruhr University Bochum, Universitätsstr. 150, D-44780 Bochum, Germany
| | - Michelle Groemmke
- Department of Zoology and Neurobiology, ND7/31, Ruhr University Bochum, Universitätsstr. 150, D-44780 Bochum, Germany
| | - Stefan Herlitze
- Department of Zoology and Neurobiology, ND7/31, Ruhr University Bochum, Universitätsstr. 150, D-44780 Bochum, Germany
| |
Collapse
|
23
|
Aikawa T, Mogushi K, Iijima-Tsutsui K, Ishikawa K, Sakurai M, Tanaka H, Mizusawa H, Watase K. Loss of MyD88 alters neuroinflammatory response and attenuates early Purkinje cell loss in a spinocerebellar ataxia type 6 mouse model. Hum Mol Genet 2015; 24:4780-91. [PMID: 26034136 PMCID: PMC4527484 DOI: 10.1093/hmg/ddv202] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/26/2015] [Indexed: 11/14/2022] Open
Abstract
Spinocerebellar ataxia type 6 (SCA6) is dominantly inherited neurodegenerative disease, caused by an expansion of CAG repeat encoding a polyglutamine (PolyQ) tract in the Cav2.1 voltage-gated calcium channel. Its key pathological features include selective degeneration of the cerebellar Purkinje cells (PCs), a common target for PolyQ-induced toxicity in various SCAs. Mutant Cav2.1 confers toxicity primarily through a toxic gain-of-function mechanism; however, its molecular basis remains elusive. Here, we studied the cerebellar gene expression patterns of young Sca6-MPI(118Q/118Q) knockin (KI) mice, which expressed mutant Cav2.1 from an endogenous locus and recapitulated many phenotypic features of human SCA6. Transcriptional signatures in the MPI(118Q/118Q) mice were distinct from those in the Sca1(154Q/2Q) mice, a faithful SCA1 KI mouse model. Temporal expression profiles of the candidate genes revealed that the up-regulation of genes associated with microglial activation was initiated before PC degeneration and was augmented as the disease progressed. Histological analysis of the MPI(118Q/118Q) cerebellum showed the predominance of M1-like pro-inflammatory microglia and it was concomitant with elevated expression levels of tumor necrosis factor, interleukin-6, Toll-like receptor (TLR) 2 and 7. Genetic ablation of MyD88, a major adaptor protein conveying TLR signaling, altered expression patterns of M1/M2 microglial phenotypic markers in the MPI(118Q/118Q) cerebellum. More importantly, it ameliorated PC loss and partially rescued motor impairments in the early disease phase. These results suggest that early neuroinflammatory response may play an important role in the pathogenesis of SCA6 and its modulation could pave the way for slowing the disease progression during the early stage of the disease.
Collapse
Affiliation(s)
- Tomonori Aikawa
- Center for Brain Integration Research, Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan
| | - Kaoru Mogushi
- Department of Bioinformatics, Medical Research Institute, Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan, Center for Genomic and Regenerative Medicine, Juntendo University, Tokyo 113-0033, Japan
| | - Kumiko Iijima-Tsutsui
- Department of Bioinformatics, Medical Research Institute, Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan, Department of Social Services and Healthcare Management, International University of Health and Welfare, Tochigi 324-8501, Japan and
| | - Kinya Ishikawa
- Department of Neurology and Neurogical Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan, Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan
| | | | - Hiroshi Tanaka
- Department of Bioinformatics, Medical Research Institute, Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan
| | - Hidehiro Mizusawa
- Center for Brain Integration Research, Department of Neurology and Neurogical Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan, Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan
| | - Kei Watase
- Center for Brain Integration Research, Core Research for Evolutional Science and Technology (CREST) of the Japan Science and Technology (JST), Tokyo 102-8666, Japan,
| |
Collapse
|
24
|
Spinocerebellar ataxia type 6 protein aggregates cause deficits in motor learning and cerebellar plasticity. J Neurosci 2015; 35:8882-95. [PMID: 26063920 DOI: 10.1523/jneurosci.0891-15.2015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Spinocerebellar ataxia type 6 (SCA6) is linked to poly-glutamine (polyQ) within the C terminus (CT) of the pore-forming subunits of P/Q-type Ca(2+) channels (Cav2.1) and is characterized by CT protein aggregates found in cerebellar Purkinje cells (PCs). One hypothesis regarding SCA6 disease is that a CT fragment of the Cav2.1 channel, which is detected specifically in cytosolic and nuclear fractions in SCA6 patients, is associated with the SCA6 pathogenesis. To test this hypothesis, we expressed P/Q-type channel protein fragments from two different human CT splice variants, as predicted from SCA6 patients, in PCs of mice using viral and transgenic approaches. These splice variants represent a short (CT-short without polyQs) and a long (CT-long with 27 polyQs) CT fragment. Our results show that the different splice variants of the CTs differentially distribute within PCs, i.e., the short CTs reveal predominantly nuclear inclusions, whereas the long CTs prominently reveal both nuclear and cytoplasmic aggregates. Postnatal expression of CTs in PCs in mice reveals that only CT-long causes SCA6-like symptoms, i.e., deficits in eyeblink conditioning (EBC), ataxia, and PC degeneration. The physiological phenotypes associated specifically with the long CT fragment can be explained by an impairment of LTD and LTP at the parallel fiber-to-PC synapse and alteration in spontaneous PC activity. Thus, our results suggest that the polyQ carrying the CT fragment of the P/Q-type channel is sufficient to cause SCA6 pathogenesis in mice and identifies EBC as a new diagnostic strategy to evaluate Ca(2+) channel-mediated human diseases.
Collapse
|
25
|
Merrill MJ, Nai D, Ghosh P, Edwards NA, Hallett M, Ray-Chaudhury A. Neuropathology in a case of episodic ataxia type 4. Neuropathol Appl Neurobiol 2015; 42:296-300. [PMID: 26264377 DOI: 10.1111/nan.12262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/05/2015] [Indexed: 01/23/2023]
Affiliation(s)
- M J Merrill
- Surgical Neurology Branch, NINDS, NIH, Bethesda, USA
| | - D Nai
- Surgical Neurology Branch, NINDS, NIH, Bethesda, USA
| | - P Ghosh
- Medical Neurology Branch, NINDS, NIH, Bethesda, USA.,Department of Neurology, George Washington University Medical Faculty Associates, Washington, USA
| | - N A Edwards
- Surgical Neurology Branch, NINDS, NIH, Bethesda, USA
| | - M Hallett
- Medical Neurology Branch, NINDS, NIH, Bethesda, USA
| | | |
Collapse
|
26
|
Nishina K, Piao W, Yoshida-Tanaka K, Sujino Y, Nishina T, Yamamoto T, Nitta K, Yoshioka K, Kuwahara H, Yasuhara H, Baba T, Ono F, Miyata K, Miyake K, Seth PP, Low A, Yoshida M, Bennett CF, Kataoka K, Mizusawa H, Obika S, Yokota T. DNA/RNA heteroduplex oligonucleotide for highly efficient gene silencing. Nat Commun 2015; 6:7969. [PMID: 26258894 PMCID: PMC4918363 DOI: 10.1038/ncomms8969] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 07/01/2015] [Indexed: 12/13/2022] Open
Abstract
Antisense oligonucleotides (ASOs) are recognized therapeutic agents for the modulation of specific genes at the post-transcriptional level. Similar to any medical drugs, there are opportunities to improve their efficacy and safety. Here we develop a short DNA/RNA heteroduplex oligonucleotide (HDO) with a structure different from double-stranded RNA used for short interfering RNA and single-stranded DNA used for ASO. A DNA/locked nucleotide acid gapmer duplex with an α-tocopherol-conjugated complementary RNA (Toc-HDO) is significantly more potent at reducing the expression of the targeted mRNA in liver compared with the parent single-stranded gapmer ASO. Toc-HDO also improves the phenotype in disease models more effectively. In addition, the high potency of Toc-HDO results in a reduction of liver dysfunction observed in the parent ASO at a similar silencing effect. HDO technology offers a novel concept of therapeutic oligonucleotides, and the development of this molecular design opens a new therapeutic field.
Collapse
Affiliation(s)
- Kazutaka Nishina
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
- Section of Molecular Technology, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Wenying Piao
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
- Section of Molecular Technology, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Kie Yoshida-Tanaka
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
- Section of Molecular Technology, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Yumiko Sujino
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
- Section of Molecular Technology, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Tomoko Nishina
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
- Section of Molecular Technology, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Tsuyoshi Yamamoto
- Bioorganic Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita-shi, Osaka 565-0871, Japan
| | - Keiko Nitta
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Kotaro Yoshioka
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
- Section of Molecular Technology, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Hiroya Kuwahara
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
- Section of Molecular Technology, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Hidenori Yasuhara
- Bioorganic Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita-shi, Osaka 565-0871, Japan
| | - Takeshi Baba
- Bioorganic Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita-shi, Osaka 565-0871, Japan
| | - Fumiko Ono
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba-shi, Ibaraki 305-0003, Japan
| | - Kanjiro Miyata
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Koichi Miyake
- Department of Biochemistry and Molecular Biology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Punit P. Seth
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, California 92010, USA
| | - Audrey Low
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, California 92010, USA
| | - Masayuki Yoshida
- Department of Life Science and Medical Ethics, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - C. Frank Bennett
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, California 92010, USA
| | - Kazunori Kataoka
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Hidehiro Mizusawa
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Satoshi Obika
- Section of Molecular Technology, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
- Bioorganic Chemistry, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita-shi, Osaka 565-0871, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
- Section of Molecular Technology, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| |
Collapse
|
27
|
Ohmori H, Hara A, Ishikawa K, Mizusawa H, Ando Y. Clinical characteristics of combined cases of spinocerebellar ataxia types 6 and 31. J Neurogenet 2015; 29:80-4. [PMID: 26004545 DOI: 10.3109/01677063.2015.1054992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study reports the first family in which spinocerebellar ataxia type 6 (SCA6) and spinocerebellar ataxia type 31 (SCA31) mutations were seen. An index patient first presented to our hospital due to gait and speech disturbances. Subsequent clinical investigation of this patient and her family members revealed consistent pure cerebellar ataxia transmitted in an autosomal-dominant manner. Genetic examination unexpectedly demonstrated that two of the five affected individuals had expansions of SCA6 and SCA31, while two others had SCA31 alone and the remaining had SCA6. Clinical manifestations were more severe in individuals with combined mutations relative to those with single mutation, suggesting that the SCA6 and SCA31 mutations have a cumulative pathogenic effect.
Collapse
Affiliation(s)
- Hiroyuki Ohmori
- a Department of Neurology , Yamaga Chuo Hospital , Yamaga City, Kumamoto , Japan
| | - Akio Hara
- a Department of Neurology , Yamaga Chuo Hospital , Yamaga City, Kumamoto , Japan
| | - Kinya Ishikawa
- b Department of Neurology and Neurological Science , Tokyo Medical and Dental University , Tokyo , Japan
| | - Hidehiro Mizusawa
- b Department of Neurology and Neurological Science , Tokyo Medical and Dental University , Tokyo , Japan
| | - Yukio Ando
- c Department of Neurology , Graduate School of Medical Sciences, Kumamoto University , Kumamoto , Japan
| |
Collapse
|
28
|
Tsou WL, Hosking RR, Burr AA, Sutton JR, Ouyang M, Du X, Gomez CM, Todi SV. DnaJ-1 and karyopherin α3 suppress degeneration in a new Drosophila model of Spinocerebellar Ataxia Type 6. Hum Mol Genet 2015; 24:4385-96. [PMID: 25954029 DOI: 10.1093/hmg/ddv174] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/05/2015] [Indexed: 11/15/2022] Open
Abstract
Spinocerebellar ataxia type 6 (SCA6) belongs to the family of CAG/polyglutamine (polyQ)-dependent neurodegenerative disorders. SCA6 is caused by abnormal expansion in a CAG trinucleotide repeat within exon 47 of CACNA1A, a bicistronic gene that encodes α1A, a P/Q-type calcium channel subunit and a C-terminal protein, termed α1ACT. Expansion of the CAG/polyQ region of CACNA1A occurs within α1ACT and leads to ataxia. There are few animal models of SCA6. Here, we describe the generation and characterization of the first Drosophila melanogaster models of SCA6, which express the entire human α1ACT protein with a normal or expanded polyQ. The polyQ-expanded version of α1ACT recapitulates the progressively degenerative nature of SCA6 when expressed in various fly tissues and the presence of densely staining aggregates. Additional studies identify the co-chaperone DnaJ-1 as a potential therapeutic target for SCA6. Expression of DnaJ-1 potently suppresses α1ACT-dependent degeneration and lethality, concomitant with decreased aggregation and reduced nuclear localization of the pathogenic protein. Mutating the nuclear importer karyopherin α3 also leads to reduced toxicity from pathogenic α1ACT. Little is known about the steps leading to degeneration in SCA6 and the means to protect neurons in this disease are lacking. Invertebrate animal models of SCA6 can expand our understanding of molecular sequelae related to degeneration in this disorder and lead to the rapid identification of cellular components that can be targeted to treat it.
Collapse
Affiliation(s)
| | | | - Aaron A Burr
- Department of Pharmacology, Cancer Biology Graduate Program and
| | | | | | - Xiaofei Du
- Department of Neurology, University of Chicago School of Medicine, Chicago, IL 60637, USA
| | - Christopher M Gomez
- Department of Neurology, University of Chicago School of Medicine, Chicago, IL 60637, USA
| | - Sokol V Todi
- Department of Pharmacology, Cancer Biology Graduate Program and Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA and
| |
Collapse
|
29
|
Giunti P, Mantuano E, Frontali M, Veneziano L. Molecular mechanism of Spinocerebellar Ataxia type 6: glutamine repeat disorder, channelopathy and transcriptional dysregulation. The multifaceted aspects of a single mutation. Front Cell Neurosci 2015; 9:36. [PMID: 25762895 PMCID: PMC4329791 DOI: 10.3389/fncel.2015.00036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/21/2015] [Indexed: 11/23/2022] Open
Abstract
Spinocerebellar Ataxia type 6 (SCA6) is an autosomal dominant neurodegenerative disease characterized by late onset, slowly progressive, mostly pure cerebellar ataxia. It is one of three allelic disorders associated to CACNA1A gene, coding for the Alpha1 A subunit of P/Q type calcium channel Cav2.1 expressed in the brain, particularly in the cerebellum. The other two disorders are Episodic Ataxia type 2 (EA2), and Familial Hemiplegic Migraine type 1 (FHM1). These disorders show distinct phenotypes that often overlap but have different pathogenic mechanisms. EA2 and FHM1 are due to mutations causing, respectively, a loss and a gain of channel function. SCA6, instead, is associated with short expansions of a polyglutamine stretch located in the cytoplasmic C-terminal tail of the protein. This domain has a relevant role in channel regulation, as well as in transcription regulation of other neuronal genes; thus the SCA6 CAG repeat expansion results in complex pathogenic molecular mechanisms reflecting the complex Cav2.1 C-terminus activity. We will provide a short review for an update on the SCA6 molecular mechanism.
Collapse
Affiliation(s)
- Paola Giunti
- Laboratory of Neurogenetics, Department of Molecular Neuroscience, UCL Institute of Neurology London, UK
| | - Elide Mantuano
- Laboratory of Neurogenetics, Institute of Translational Pharmacology, National Research Council of Italy Rome, Italy
| | - Marina Frontali
- Laboratory of Neurogenetics, Institute of Translational Pharmacology, National Research Council of Italy Rome, Italy
| | - Liana Veneziano
- Laboratory of Neurogenetics, Institute of Translational Pharmacology, National Research Council of Italy Rome, Italy
| |
Collapse
|
30
|
Ota K, Obayashi M, Ozaki K, Ichinose S, Kakita A, Tada M, Takahashi H, Ando N, Eishi Y, Mizusawa H, Ishikawa K. Relocation of p25α/tubulin polymerization promoting protein from the nucleus to the perinuclear cytoplasm in the oligodendroglia of sporadic and COQ2 mutant multiple system atrophy. Acta Neuropathol Commun 2014; 2:136. [PMID: 25208467 PMCID: PMC4172786 DOI: 10.1186/s40478-014-0136-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 08/31/2014] [Indexed: 02/08/2023] Open
Abstract
p25α/tubulin polymerization promoting protein (TPPP) is an oligodendroglial protein that plays crucial roles including myelination, and the stabilization of microtubules. In multiple system atrophy (MSA), TPPP is suggested to relocate from the myelin sheath to the oligodendroglial cell body, before the formation of glial cytoplasmic inclusions (GCIs), the pathologic hallmark of MSA. However, much is left unknown about the re-distribution of TPPP in MSA. We generated new antibodies against the N- and C-terminus of TPPP, and analyzed control and MSA brains, including the brain of a familial MSA patient carrying homozygous mutations in the coenzyme Q2 gene (COQ2). In control brain tissues, TPPP was localized not only in the cytoplasmic component of the oligodendroglia including perinuclear cytoplasm and peripheral processes in the white matter, but also in the nucleus of a fraction (62.4%) of oligodendroglial cells. Immunoelectron microscopic analysis showed TPPP in the nucleus and mitochondrial membrane of normal oligodendroglia, while western blot also supported its nuclear and mitochondrial existence. In MSA, the prevalence of nuclear TPPP was 48.6% in the oligodendroglia lacking GCIs, whereas it was further decreased to 19.6% in the oligodendroglia with phosphorylated α-synuclein (pα-syn)-positive GCIs, both showing a significant decrease compared to controls (62.4%). In contrast, TPPP accumulated in the perinuclear cytoplasm where mitochondrial membrane (TOM20 and cytochrome C) and fission (DRP1) proteins were often immunoreactive. We conclude that in MSA-oligodendroglia, TPPP is reduced, not only in the peripheral cytoplasm, but also in the nucleus and relocated to the perinuclear cytoplasm.
Collapse
|
31
|
Mutational Consequences of Aberrant Ion Channels in Neurological Disorders. J Membr Biol 2014; 247:1083-127. [DOI: 10.1007/s00232-014-9716-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 07/25/2014] [Indexed: 12/25/2022]
|
32
|
Mohan RD, Abmayr SM, Workman JL. The expanding role for chromatin and transcription in polyglutamine disease. Curr Opin Genet Dev 2014; 26:96-104. [PMID: 25108806 DOI: 10.1016/j.gde.2014.06.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 06/22/2014] [Accepted: 06/25/2014] [Indexed: 11/28/2022]
Abstract
Nine genetic diseases arise from expansion of CAG repeats in seemingly unrelated genes. They are referred to as polyglutamine (polyQ) diseases due to the presence of elongated glutamine tracts in the corresponding proteins. The pathologic consequences of polyQ expansion include progressive spinal, cerebellar, and neural degeneration. These pathologies are not identical, however, suggesting that disruption of protein-specific functions is crucial to establish and maintain each disease. A closer examination of protein function reveals that several act as regulators of gene expression. Here we examine the roles these proteins play in regulating gene expression, discuss how polyQ expansion may disrupt these functions to cause disease, and speculate on the neural specificity of perturbing ubiquitous gene regulators.
Collapse
Affiliation(s)
- Ryan D Mohan
- Stowers Institute for Medical Research, 1000 E 50th St., Kansas City, MO 64110, USA
| | - Susan M Abmayr
- Stowers Institute for Medical Research, 1000 E 50th St., Kansas City, MO 64110, USA.
| | - Jerry L Workman
- Stowers Institute for Medical Research, 1000 E 50th St., Kansas City, MO 64110, USA.
| |
Collapse
|
33
|
Du X, Wang J, Zhu H, Rinaldo L, Lamar KM, Palmenberg AC, Hansel C, Gomez CM. Second cistron in CACNA1A gene encodes a transcription factor mediating cerebellar development and SCA6. Cell 2013; 154:118-33. [PMID: 23827678 PMCID: PMC3939801 DOI: 10.1016/j.cell.2013.05.059] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 02/05/2013] [Accepted: 05/31/2013] [Indexed: 12/18/2022]
Abstract
The CACNA1A gene, encoding the voltage-gated calcium channel subunit α1A, is involved in pre- and postsynaptic Ca(2+) signaling, gene expression, and several genetic neurological disorders. We found that CACNA1A coordinates gene expression using a bicistronic mRNA bearing a cryptic internal ribosomal entry site (IRES). The first cistron encodes the well-characterized α1A subunit. The second expresses a transcription factor, α1ACT, which coordinates expression of a program of genes involved in neural and Purkinje cell development. α1ACT also contains the polyglutamine (polyQ) tract that, when expanded, causes spinocerebellar ataxia type 6 (SCA6). When expressed as an independent polypeptide, α1ACT-bearing an expanded polyQ tract-lacks transcription factor function and neurite outgrowth properties, causes cell death in culture, and leads to ataxia and cerebellar atrophy in transgenic mice. Suppression of CACNA1A IRES function in SCA6 may be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Xiaofei Du
- Department of Neurology, University of Chicago, IL 60637, USA
| | - Jun Wang
- Department of Neurology, University of Chicago, IL 60637, USA
| | - Haipeng Zhu
- Department of Neurology, University of Chicago, IL 60637, USA
| | - Lorenzo Rinaldo
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Kay-Marie Lamar
- Department of Neurology, University of Chicago, IL 60637, USA
| | - Ann C. Palmenberg
- Institute for Molecular Virology, University of Wisconsin-Madison, WI 53706, USA
| | - Christian Hansel
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
34
|
Yamaura H, Hirai H, Yanagihara D. Postural dysfunction in a transgenic mouse model of spinocerebellar ataxia type 3. Neuroscience 2013; 243:126-35. [DOI: 10.1016/j.neuroscience.2013.03.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 03/07/2013] [Accepted: 03/24/2013] [Indexed: 12/23/2022]
|
35
|
Niimi Y, Takahashi M, Sugawara E, Umeda S, Obayashi M, Sato N, Ishiguro T, Higashi M, Eishi Y, Mizusawa H, Ishikawa K. Abnormal RNA structures (RNA foci) containing a penta-nucleotide repeat (UGGAA)nin the Purkinje cell nucleus is associated with spinocerebellar ataxia type 31 pathogenesis. Neuropathology 2013; 33:600-11. [DOI: 10.1111/neup.12032] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 02/12/2013] [Accepted: 02/13/2013] [Indexed: 11/27/2022]
Affiliation(s)
- Yusuke Niimi
- Department of Neurology and Neurological Science; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Makoto Takahashi
- Department of Neurology and Neurological Science; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Emiko Sugawara
- Department of Pathology; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Shigeaki Umeda
- Department of Pathology; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Masato Obayashi
- Department of Neurology and Neurological Science; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Nozomu Sato
- Department of Neurology and Neurological Science; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Taro Ishiguro
- Department of Neurology and Neurological Science; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Miwa Higashi
- Department of Neurology and Neurological Science; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Yoshinobu Eishi
- Department of Pathology; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Hidehiro Mizusawa
- Department of Neurology and Neurological Science; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - Kinya Ishikawa
- Department of Neurology and Neurological Science; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| |
Collapse
|
36
|
Takahashi M, Obayashi M, Ishiguro T, Sato N, Niimi Y, Ozaki K, Mogushi K, Mahmut Y, Tanaka H, Tsuruta F, Dolmetsch R, Yamada M, Takahashi H, Kato T, Mori O, Eishi Y, Mizusawa H, Ishikawa K. Cytoplasmic location of α1A voltage-gated calcium channel C-terminal fragment (Cav2.1-CTF) aggregate is sufficient to cause cell death. PLoS One 2013; 8:e50121. [PMID: 23505410 PMCID: PMC3591409 DOI: 10.1371/journal.pone.0050121] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 10/17/2012] [Indexed: 01/20/2023] Open
Abstract
The human α1A voltage-dependent calcium channel (Cav2.1) is a pore-forming essential subunit embedded in the plasma membrane. Its cytoplasmic carboxyl(C)-tail contains a small poly-glutamine (Q) tract, whose length is normally 4∼19 Q, but when expanded up to 20∼33Q, the tract causes an autosomal-dominant neurodegenerative disorder, spinocerebellar ataxia type 6 (SCA6). A recent study has shown that a 75-kDa C-terminal fragment (CTF) containing the polyQ tract remains soluble in normal brains, but becomes insoluble mainly in the cytoplasm with additional localization to the nuclei of human SCA6 Purkinje cells. However, the mechanism by which the CTF aggregation leads to neurodegeneration is completely elusive, particularly whether the CTF exerts more toxicity in the nucleus or in the cytoplasm. We tagged recombinant (r)CTF with either nuclear-localization or nuclear-export signal, created doxycyclin-inducible rat pheochromocytoma (PC12) cell lines, and found that the CTF is more toxic in the cytoplasm than in the nucleus, the observations being more obvious with Q28 (disease range) than with Q13 (normal-length). Surprisingly, the CTF aggregates co-localized both with cAMP response element-binding protein (CREB) and phosphorylated-CREB (p-CREB) in the cytoplasm, and Western blot analysis showed that the quantity of CREB and p-CREB were both decreased in the nucleus when the rCTF formed aggregates in the cytoplasm. In human brains, polyQ aggregates also co-localized with CREB in the cytoplasm of SCA6 Purkinje cells, but not in other conditions. Collectively, the cytoplasmic Cav2.1-CTF aggregates are sufficient to cause cell death, and one of the pathogenic mechanisms may be abnormal CREB trafficking in the cytoplasm and reduced CREB and p-CREB levels in the nuclei.
Collapse
Affiliation(s)
- Makoto Takahashi
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Masato Obayashi
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Taro Ishiguro
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Nozomu Sato
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Yusuke Niimi
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Kokoro Ozaki
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Kaoru Mogushi
- Information Center for Medical Sciences, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Yasen Mahmut
- Information Center for Medical Sciences, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Tanaka
- Information Center for Medical Sciences, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Fuminori Tsuruta
- Department of Neurobiology, Stanford University School of Medicine, Fairchild Research Building, Palo Alto, California, United States of America
| | - Ricardo Dolmetsch
- Department of Neurobiology, Stanford University School of Medicine, Fairchild Research Building, Palo Alto, California, United States of America
| | - Mitsunori Yamada
- Department of Pathology, Pathological Neuroscience Branch, Brain Research Institute, Niigata University, Asahi-machi-dori, Niigata, Japan
- Department of Clinical Research, National Hospital Organization, Saigata National Hospital, Saigata, Ohgata-ku, Johetsu-City, Niigata, Japan
| | - Hitoshi Takahashi
- Department of Pathology, Pathological Neuroscience Branch, Brain Research Institute, Niigata University, Asahi-machi-dori, Niigata, Japan
| | - Takeo Kato
- Department of Neurology, Hematology, Metabolism, Endocrinology and Diabetology, Yamagata University Faculty of Medicine, Iida-Nishi, Yamagata, Japan
| | - Osamu Mori
- Department of Internal Medicine and Neurology, Hatsuishi Hospital, Nishihara, Kashiwa, Chiba, Japan
| | - Yoshinobu Eishi
- Department of Pathology, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Hidehiro Mizusawa
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
| | - Kinya Ishikawa
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
37
|
Rüb U, Schöls L, Paulson H, Auburger G, Kermer P, Jen JC, Seidel K, Korf HW, Deller T. Clinical features, neurogenetics and neuropathology of the polyglutamine spinocerebellar ataxias type 1, 2, 3, 6 and 7. Prog Neurobiol 2013; 104:38-66. [PMID: 23438480 DOI: 10.1016/j.pneurobio.2013.01.001] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 01/22/2013] [Accepted: 01/31/2013] [Indexed: 12/18/2022]
Abstract
The spinocerebellar ataxias type 1 (SCA1), 2 (SCA2), 3 (SCA3), 6 (SCA6) and 7 (SCA7) are genetically defined autosomal dominantly inherited progressive cerebellar ataxias (ADCAs). They belong to the group of CAG-repeat or polyglutamine diseases and share pathologically expanded and meiotically unstable glutamine-encoding CAG-repeats at distinct gene loci encoding elongated polyglutamine stretches in the disease proteins. In recent years, progress has been made in the understanding of the pathogenesis of these currently incurable diseases: Identification of underlying genetic mechanisms made it possible to classify the different ADCAs and to define their clinical and pathological features. Furthermore, advances in molecular biology yielded new insights into the physiological and pathophysiological role of the gene products of SCA1, SCA2, SCA3, SCA6 and SCA7 (i.e. ataxin-1, ataxin-2, ataxin-3, α-1A subunit of the P/Q type voltage-dependent calcium channel, ataxin-7). In the present review we summarize our current knowledge about the polyglutamine ataxias SCA1, SCA2, SCA3, SCA6 and SCA7 and compare their clinical and electrophysiological features, genetic and molecular biological background, as well as their brain pathologies. Furthermore, we provide an overview of the structure, interactions and functions of the different disease proteins. On the basis of these comprehensive data, similarities, differences and possible disease mechanisms are discussed.
Collapse
Affiliation(s)
- Udo Rüb
- Dr. Senckenberg Chronomedical Institute, Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt/Main, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Camero S, Benítez MJ, Jiménez JS. Anomalous Protein–DNA Interactions Behind Neurological Disorders. PROTEIN-NUCLEIC ACIDS INTERACTIONS 2013; 91:37-63. [DOI: 10.1016/b978-0-12-411637-5.00002-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Development of Purkinje cell degeneration in a knockin mouse model reveals lysosomal involvement in the pathogenesis of SCA6. Proc Natl Acad Sci U S A 2012; 109:17693-8. [PMID: 23054835 DOI: 10.1073/pnas.1212786109] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Spinocerebellar ataxia type 6 (SCA6) is a neurodegenerative disease caused by the expansion of a polyglutamine tract in the Ca(v)2.1 voltage-gated calcium channel. To elucidate how the expanded polyglutamine tract in this plasma membrane protein causes the disease, we created a unique knockin mouse model that modestly overexpressed the mutant transcripts under the control of an endogenous promoter (MPI-118Q). MPI-118Q mice faithfully recapitulated many features of SCA6, including selective Purkinje cell degeneration. Surprisingly, analysis of inclusion formation in the mutant Purkinje cells indicated the lysosomal localization of accumulated mutant Ca(v)2.1 channels in the absence of autophagic response. The lack of cathepsin B, a major lysosomal cysteine proteinase, exacerbated the loss of Purkinje cells and was accompanied by an acceleration of inclusion formation in this model. Thus, the pathogenic mechanism of SCA6 involves the endolysosomal degradation pathway, and unique pathological features of this model further illustrate the pivotal role of protein context in the pathogenesis of polyglutamine diseases.
Collapse
|
40
|
Seidel K, Siswanto S, Brunt ERP, den Dunnen W, Korf HW, Rüb U. Brain pathology of spinocerebellar ataxias. Acta Neuropathol 2012; 124:1-21. [PMID: 22684686 DOI: 10.1007/s00401-012-1000-x] [Citation(s) in RCA: 291] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/25/2012] [Accepted: 05/25/2012] [Indexed: 12/22/2022]
Abstract
The autosomal dominant cerebellar ataxias (ADCAs) represent a heterogeneous group of neurodegenerative diseases with progressive ataxia and cerebellar degeneration. The current classification of this disease group is based on the underlying genetic defects and their typical disease courses. According to this categorization, ADCAs are divided into the spinocerebellar ataxias (SCAs) with a progressive disease course, and the episodic ataxias (EA) with episodic occurrences of ataxia. The prominent disease symptoms of the currently known and genetically defined 31 SCA types result from damage to the cerebellum and interconnected brain grays and are often accompanied by more specific extra-cerebellar symptoms. In the present review, we report the genetic and clinical background of the known SCAs and present the state of neuropathological investigations of brain tissue from SCA patients in the final disease stages. Recent findings show that the brain is commonly seriously affected in the polyglutamine SCAs (i.e. SCA1, SCA2, SCA3, SCA6, SCA7, and SCA17) and that the patterns of brain damage in these diseases overlap considerably in patients suffering from advanced disease stages. In the more rarely occurring non-polyglutamine SCAs, post-mortem neuropathological data currently are scanty and investigations have been primarily performed in vivo by means of MRI brain imaging. Only a minority of SCAs exhibit symptoms and degenerative patterns allowing for a clear and unambiguous diagnosis of the disease, e.g. retinal degeneration in SCA7, tau aggregation in SCA11, dentate calcification in SCA20, protein depositions in the Purkinje cell layer in SCA31, azoospermia in SCA32, and neurocutaneous phenotype in SCA34. The disease proteins of polyglutamine ataxias and some non-polyglutamine ataxias aggregate as cytoplasmic or intranuclear inclusions and serve as morphological markers. Although inclusions may impair axonal transport, bind transcription factors, and block protein quality control, detailed molecular and pathogenetic consequences remain to be determined.
Collapse
Affiliation(s)
- Kay Seidel
- Dr. Senckenbergisches Chronomedizinisches Institut, Goethe University, Theodor-Stern-Kai 7, 60950, Frankfurt/Main, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Takahashi M, Ishikawa K, Sato N, Obayashi M, Niimi Y, Ishiguro T, Yamada M, Toyoshima Y, Takahashi H, Kato T, Takao M, Murayama S, Mori O, Eishi Y, Mizusawa H. Reduced brain-derived neurotrophic factor (BDNF) mRNA expression and presence of BDNF-immunoreactive granules in the spinocerebellar ataxia type 6 (SCA6) cerebellum. Neuropathology 2012; 32:595-603. [DOI: 10.1111/j.1440-1789.2012.01302.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Nestor CE, Monckton DG. Correlation of inter-locus polyglutamine toxicity with CAG•CTG triplet repeat expandability and flanking genomic DNA GC content. PLoS One 2011; 6:e28260. [PMID: 22163004 PMCID: PMC3232215 DOI: 10.1371/journal.pone.0028260] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 11/04/2011] [Indexed: 11/19/2022] Open
Abstract
Dynamic expansions of toxic polyglutamine (polyQ)-encoding CAG repeats in ubiquitously expressed, but otherwise unrelated, genes cause a number of late-onset progressive neurodegenerative disorders, including Huntington disease and the spinocerebellar ataxias. As polyQ toxicity in these disorders increases with repeat length, the intergenerational expansion of unstable CAG repeats leads to anticipation, an earlier age-at-onset in successive generations. Crucially, disease associated alleles are also somatically unstable and continue to expand throughout the lifetime of the individual. Interestingly, the inherited polyQ length mediating a specific age-at-onset of symptoms varies markedly between disorders. It is widely assumed that these inter-locus differences in polyQ toxicity are mediated by protein context effects. Previously, we demonstrated that the tendency of expanded CAG•CTG repeats to undergo further intergenerational expansion (their 'expandability') also differs between disorders and these effects are strongly correlated with the GC content of the genomic flanking DNA. Here we show that the inter-locus toxicity of the expanded polyQ tracts of these disorders also correlates with both the expandability of the underlying CAG repeat and the GC content of the genomic DNA flanking sequences. Inter-locus polyQ toxicity does not correlate with properties of the mRNA or protein sequences, with polyQ location within the gene or protein, or steady state transcript levels in the brain. These data suggest that the observed inter-locus differences in polyQ toxicity are not mediated solely by protein context effects, but that genomic context is also important, an effect that may be mediated by modifying the rate at which somatic expansion of the DNA delivers proteins to their cytotoxic state.
Collapse
Affiliation(s)
- Colm E Nestor
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
| | | |
Collapse
|
43
|
Tsou WL, Soong BW, Paulson HL, Rodríguez-Lebrón E. Splice isoform-specific suppression of the Cav2.1 variant underlying spinocerebellar ataxia type 6. Neurobiol Dis 2011; 43:533-42. [PMID: 21550405 DOI: 10.1016/j.nbd.2011.04.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 04/08/2011] [Accepted: 04/21/2011] [Indexed: 01/23/2023] Open
Abstract
Spinocerebellar ataxia type 6 (SCA6) is an inherited neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the Ca(V)2.1 voltage-gated calcium channel subunit (CACNA1A). There is currently no treatment for this debilitating disorder and thus a pressing need to develop preventative therapies. RNA interference (RNAi) has proven effective at halting disease progression in several models of spinocerebellar ataxia (SCA), including SCA types 1 and 3. However, in SCA6 and other dominantly inherited neurodegenerative disorders, RNAi-based strategies that selectively suppress expression of mutant alleles may be required. Using a Ca(V)2.1 mini-gene reporter system, we found that pathogenic CAG expansions in Ca(V)2.1 enhance splicing activity at the 3'end of the transcript, leading to a CAG repeat length-dependent increase in the levels of a polyQ-encoding Ca(V)2.1 mRNA splice isoform and the resultant disease protein. Taking advantage of this molecular phenomenon, we developed a novel splice isoform-specific (SIS)-RNAi strategy that selectively targets the polyQ-encoding Ca(V)2.1 splice variant. Selective suppression of transiently expressed and endogenous polyQ-encoding Ca(V)2.1 splice variants was achieved in a variety of cell-based models including a human neuronal cell line, using a new artificial miRNA-like delivery system. Moreover, the efficacy of gene silencing correlated with effective intracellular recognition and processing of SIS-RNAi miRNA mimics. These results lend support to the preclinical development of SIS-RNAi as a potential therapy for SCA6 and other dominantly inherited diseases.
Collapse
Affiliation(s)
- Wei-Ling Tsou
- Institute of Neuroscience, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | | | | | | |
Collapse
|