1
|
Borbolla-Jiménez FV, García-Aguirre IA, Del Prado-Audelo ML, Hernández-Hernández O, Cisneros B, Leyva-Gómez G, Magaña JJ. Development of a Polymeric Pharmacological Nanocarrier System as a Potential Therapy for Spinocerebellar Ataxia Type 7. Cells 2023; 12:2735. [PMID: 38067163 PMCID: PMC10706302 DOI: 10.3390/cells12232735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Spinocerebellar ataxia type 7 (SCA7) is an autosomal-dominant inherited disease characterized by progressive ataxia and retinal degeneration. SCA7 belongs to a group of neurodegenerative diseases caused by an expanded CAG repeat in the disease-causing gene, resulting in aberrant polyglutamine (polyQ) protein synthesis. PolyQ ataxin-7 is prone to aggregate in intracellular inclusions, perturbing cellular processes leading to neuronal death in specific regions of the central nervous system (CNS). Currently, there is no treatment for SCA7; however, a promising approach successfully applied to other polyQ diseases involves the clearance of polyQ protein aggregates through pharmacological activation of autophagy. Nonetheless, the blood-brain barrier (BBB) poses a challenge for delivering drugs to the CNS, limiting treatment effectiveness. This study aimed to develop a polymeric nanocarrier system to deliver therapeutic agents across the BBB into the CNS. We prepared poly(lactic-co-glycolic acid) nanoparticles (NPs) modified with Poloxamer188 and loaded with rapamycin to enable NPs to activate autophagy. We demonstrated that these rapamycin-loaded NPs were successfully taken up by neuronal and glial cells, demonstrating high biocompatibility without adverse effects. Remarkably, rapamycin-loaded NPs effectively cleared mutant ataxin-7 aggregates in a SCA7 glial cell model, highlighting their potential as a therapeutic approach to fight SCA7 and other polyQ diseases.
Collapse
Affiliation(s)
- Fabiola V. Borbolla-Jiménez
- Laboratorio de Medicina Genómica, Departamento de Genética (CENIAQ), Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra (INR-LGII), Ciudad de México 14389, Mexico; (F.V.B.-J.); (O.H.-H.)
- Programa de Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - Ian A. García-Aguirre
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Ciudad de México 14380, Mexico; (I.A.G.-A.); (M.L.D.P.-A.)
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México 07360, Mexico;
| | - María Luisa Del Prado-Audelo
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Ciudad de México 14380, Mexico; (I.A.G.-A.); (M.L.D.P.-A.)
| | - Oscar Hernández-Hernández
- Laboratorio de Medicina Genómica, Departamento de Genética (CENIAQ), Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra (INR-LGII), Ciudad de México 14389, Mexico; (F.V.B.-J.); (O.H.-H.)
| | - Bulmaro Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados (CINVESTAV-IPN), Ciudad de México 07360, Mexico;
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Jonathan J. Magaña
- Laboratorio de Medicina Genómica, Departamento de Genética (CENIAQ), Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra (INR-LGII), Ciudad de México 14389, Mexico; (F.V.B.-J.); (O.H.-H.)
- Programa de Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Ciudad de México 14380, Mexico; (I.A.G.-A.); (M.L.D.P.-A.)
| |
Collapse
|
2
|
Jain M, Patil N, Abdi G, Abbasi Tarighat M, Mohammed A, Ahmad Mohd Zain MR, Goh KW. Mechanistic Insights and Potential Therapeutic Approaches in PolyQ Diseases via Autophagy. Biomedicines 2023; 11:biomedicines11010162. [PMID: 36672670 PMCID: PMC9856063 DOI: 10.3390/biomedicines11010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 01/11/2023] Open
Abstract
Polyglutamine diseases are a group of congenital neurodegenerative diseases categorized with genomic abnormalities in the expansion of CAG triplet repeats in coding regions of specific disease-related genes. Protein aggregates are the toxic hallmark for polyQ diseases and initiate neuronal death. Autophagy is a catabolic process that aids in the removal of damaged organelles or toxic protein aggregates, a process required to maintain cellular homeostasis that has the potential to fight against neurodegenerative diseases, but this pathway gets affected under diseased conditions, as there is a direct impact on autophagy-related gene expression. The increase in the accumulation of autophagy vesicles reported in neurodegenerative diseases was due to an increase in autophagy or may have been due to a decrease in autophagy flux. These reports suggested that there is a contribution of autophagy in the pathology of diseases and regulation in the process of autophagy. It was demonstrated in various disease models of polyQ diseases that autophagy upregulation by using modulators can enhance the dissolution of toxic aggregates and delay disease progression. In this review, interaction of the autophagy pathway with polyQ diseases was analyzed, and a therapeutic approach with autophagy inducing drugs was established for disease pathogenesis.
Collapse
Affiliation(s)
- Mukul Jain
- Department of Lifesciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India
- Lab 209 Cell and Developmental Biology Lab, Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Nil Patil
- Department of Lifesciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India
- Lab 209 Cell and Developmental Biology Lab, Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Gholamreza Abdi
- Department of Biotechnology, Persian Gulf Research Institute, Persian Gulf University, Bushehr, 75169, Iran
- Correspondence: (G.A.); (M.R.A.M.Z.); (K.W.G.)
| | - Maryam Abbasi Tarighat
- Faculty of Nano and Bio Science and Technology, Persian Gulf University, Bushehr 75169, Iran
| | - Arifullah Mohammed
- Department of Agriculture, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Malaysia
| | - Muhammad Rajaei Ahmad Mohd Zain
- Department of Orthopaedics, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Correspondence: (G.A.); (M.R.A.M.Z.); (K.W.G.)
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
- Correspondence: (G.A.); (M.R.A.M.Z.); (K.W.G.)
| |
Collapse
|
3
|
Saha G, Ghosh S, Dubey VK, Saudagar P. Gene Alterations Induced by Glutamine (Q) Encoding CAG Repeats Associated with Neurodegeneration. Methods Mol Biol 2023; 2575:3-23. [PMID: 36301468 DOI: 10.1007/978-1-0716-2716-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Several studies have been reported linking the role of polyglutamine (polyQ) disease-associated proteins with altered gene regulation induced by an unstable trinucleotide (CAG) repeat. Owing to their dynamic nature of expansion, these DNA repeats form secondary structures interfering with the normal cellular mechanisms like replication and transcription and, thereby, have become the underlying cause of numerous neurodegenerative disorders involving mental retardation and/or muscular or neuronal degeneration. Despite the widespread expression of the disease-causing protein, specific subsets of neurons are susceptible to specific patterns of inheritance and clinical symptoms. Although this cell-type selectivity is still elusive and less understood, it has been found that aberrant transcriptional regulation is one of the primary causes of polyQ diseases where the functions of histone-modifying complexes are disrupted. Besides, epigenetic modifications play a critical role in the pathogenesis of these diseases. In this chapter, we will be delving into how these polyQ repeats induce the self-assembly and aggregation of altered carrier proteins based on gene alterations, causing neuronal toxicity and cellular deaths. Besides, genomic instability in CAG repeats due to altered chromatin-related enzymes will be highlighted, along with epigenetic changes present in many polyQ disorders. Understanding the underlying molecular mechanisms in the root cause of these disorders will culminate in identifying therapeutic approaches for the treatment of these neurodegenerative disorders.
Collapse
Affiliation(s)
- Gundappa Saha
- Department of Basic & Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sukanya Ghosh
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology, Warangal, Telangana, India.
| |
Collapse
|
4
|
Niss F, Zaidi W, Hallberg E, Ström AL. Polyglutamine expanded Ataxin-7 induces DNA damage and alters FUS localization and function. Mol Cell Neurosci 2020; 110:103584. [PMID: 33338633 DOI: 10.1016/j.mcn.2020.103584] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 01/20/2023] Open
Abstract
Polyglutamine (polyQ) diseases, such as Spinocerebellar ataxia type 7 (SCA7), are caused by expansions of polyQ repeats in disease specific proteins. The sequestration of vital proteins into aggregates formed by polyQ proteins is believed to be a common pathological mechanism in these disorders. The RNA-binding protein FUS has been observed in polyQ aggregates, though if disruption of this protein plays a role in the neuronal dysfunction in SCA7 or other polyQ diseases remains unclear. We therefore analysed FUS localisation and function in a stable inducible PC12 cell model expressing the SCA7 polyQ protein ATXN7. We found that there was a high degree of FUS sequestration, which was associated with a more cytoplasmic FUS localisation, as well as a decreased expression of FUS regulated mRNAs. In contrast, the role of FUS in the formation of γH2AX positive DNA damage foci was unaffected. In fact, a statistical increase in the number of γH2AX foci, as well as an increased trend of single and double strand DNA breaks, detected by comet assay, could be observed in mutant ATXN7 cells. These results were further corroborated by a clear trend towards increased DNA damage in SCA7 patient fibroblasts. Our findings suggest that both alterations in the RNA regulatory functions of FUS, and increased DNA damage, may contribute to the pathology of SCA7.
Collapse
Affiliation(s)
- Frida Niss
- Stockholm University, Department of Biochemistry and Biophysics, Svante Arrhenius väg 16C, 10691 Stockholm, Sweden
| | - Wajiha Zaidi
- Stockholm University, Department of Biochemistry and Biophysics, Svante Arrhenius väg 16C, 10691 Stockholm, Sweden
| | - Einar Hallberg
- Stockholm University, Department of Biochemistry and Biophysics, Svante Arrhenius väg 16C, 10691 Stockholm, Sweden
| | - Anna-Lena Ström
- Stockholm University, Department of Biochemistry and Biophysics, Svante Arrhenius väg 16C, 10691 Stockholm, Sweden.
| |
Collapse
|
5
|
Proteasome Subunits Involved in Neurodegenerative Diseases. Arch Med Res 2020; 52:1-14. [PMID: 32962866 DOI: 10.1016/j.arcmed.2020.09.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/25/2020] [Accepted: 09/04/2020] [Indexed: 12/29/2022]
Abstract
The ubiquitin-proteasome system is the major pathway for the maintenance of protein homeostasis. Its inhibition causes accumulation of ubiquitinated proteins; this accumulation has been associated with several of the most common neurodegenerative diseases. Several genetic factors have been identified for most neurodegenerative diseases, however, most cases are considered idiopathic, thus making the study of the mechanisms of protein accumulation a relevant field of research. It is often mentioned that the biggest risk factor for neurodegenerative diseases is aging, and several groups have reported an age-related alteration of the expression of some of the 26S proteasome subunits and a reduction of its activity. Proteasome subunits interact with proteins that are known to accumulate in neurodegenerative diseases such as α-synuclein in Parkinson's, tau in Alzheimer's, and huntingtin in Huntington's diseases. These interactions have been explored for several years, but only until recently, we are beginning to understand them. In this review, we discuss the known interactions, the underlying patterns, and the phenotypes associated with the 26S proteasome subunits in the etiology and progression of neurodegenerative diseases where there is evidence of proteasome involvement. Special emphasis is made in reviewing proteasome subunits that interact with proteins known to have an age-related altered expression or to be involved in neurodegenerative diseases to explore key effectors that may trigger or augment their progression. Interestingly, while the causes of age-related reduction of some of the proteasome subunits are not known, there are specific relationships between the observed neurodegenerative disease and the affected proteasome subunits.
Collapse
|
6
|
Niewiadomska-Cimicka A, Trottier Y. Molecular Targets and Therapeutic Strategies in Spinocerebellar Ataxia Type 7. Neurotherapeutics 2019; 16:1074-1096. [PMID: 31432449 PMCID: PMC6985300 DOI: 10.1007/s13311-019-00778-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a rare autosomal dominant neurodegenerative disorder characterized by progressive neuronal loss in the cerebellum, brainstem, and retina, leading to cerebellar ataxia and blindness as major symptoms. SCA7 is due to the expansion of a CAG triplet repeat that is translated into a polyglutamine tract in ATXN7. Larger SCA7 expansions are associated with earlier onset of symptoms and more severe and rapid disease progression. Here, we summarize the pathological and genetic aspects of SCA7, compile the current knowledge about ATXN7 functions, and then focus on recent advances in understanding the pathogenesis and in developing biomarkers and therapeutic strategies. ATXN7 is a bona fide subunit of the multiprotein SAGA complex, a transcriptional coactivator harboring chromatin remodeling activities, and plays a role in the differentiation of photoreceptors and Purkinje neurons, two highly vulnerable neuronal cell types in SCA7. Polyglutamine expansion in ATXN7 causes its misfolding and intranuclear accumulation, leading to changes in interactions with native partners and/or partners sequestration in insoluble nuclear inclusions. Studies of cellular and animal models of SCA7 have been crucial to unveil pathomechanistic aspects of the disease, including gene deregulation, mitochondrial and metabolic dysfunctions, cell and non-cell autonomous protein toxicity, loss of neuronal identity, and cell death mechanisms. However, a better understanding of the principal molecular mechanisms by which mutant ATXN7 elicits neurotoxicity, and how interconnected pathogenic cascades lead to neurodegeneration is needed for the development of effective therapies. At present, therapeutic strategies using nucleic acid-based molecules to silence mutant ATXN7 gene expression are under development for SCA7.
Collapse
Affiliation(s)
- Anna Niewiadomska-Cimicka
- Institute of Genetic and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR7104), Institut National de la Santé et de la Recherche Médicale (U1258), University of Strasbourg, Illkirch, France
| | - Yvon Trottier
- Institute of Genetic and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR7104), Institut National de la Santé et de la Recherche Médicale (U1258), University of Strasbourg, Illkirch, France.
| |
Collapse
|
7
|
Cortés-Pereira E, Fernández-Tajes J, Fernández-Moreno M, Vázquez-Mosquera ME, Relaño S, Ramos-Louro P, Durán-Sotuela A, Dalmao-Fernández A, Oreiro N, Blanco FJ, Rego-Pérez I. Differential Association of Mitochondrial DNA Haplogroups J and H With the Methylation Status of Articular Cartilage: Potential Role in Apoptosis and Metabolic and Developmental Processes. Arthritis Rheumatol 2019; 71:1191-1200. [PMID: 30747498 DOI: 10.1002/art.40857] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/07/2019] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To analyze the influence of mitochondrial genome variation on the DNA methylome of articular cartilage. METHODS DNA methylation profiling was performed using data deposited in the NCBI Gene Expression Omnibus database (accession no. GSE43269). Data were obtained for 14 cartilage samples from subjects with haplogroup J and 20 cartilage samples from subjects with haplogroup H. Subsequent validation was performed in an independent subset of 7 subjects with haplogroup J and 9 with haplogroup H by RNA-seq. Correlated genes were validated by real-time polymerase chain reaction in an independent cohort of 12 subjects with haplogroup J and 12 with haplogroup H. Appropriate analyses were performed using R Bioconductor and qBasePlus software, and gene ontology analysis was conducted using DAVID version 6.8. RESULTS DNA methylation profiling revealed 538 differentially methylated loci, while whole-transcriptome profiling identified 2,384 differentially expressed genes, between cartilage samples from subjects with haplogroup H and those with haplogroup J. Seventeen genes showed an inverse correlation between methylation and expression. In terms of gene ontology, differences in correlations between methylation and expression were also detected between cartilage from subjects with haplogroup H and those with haplogroup J, highlighting a significantly enhanced apoptotic process in cartilage from subjects with haplogroup H (P = 0.007 for methylation and P = 0.019 for expression) and repressed apoptotic process in cartilage from subjects with haplogroup J (P = 0.021 for methylation), as well as a significant enrichment of genes related to metabolic processes (P = 1.93 × 10-4 for methylation and P = 6.79 x 10-4 for expression) and regulation of gene expression (P = 0.012 for methylation) in cartilage from subjects with haplogroup H, and to developmental processes (P = 0.015 for methylation and P = 8.25 x 10-12 for expression) in cartilage from subjects with haplogroup J. CONCLUSION Mitochondrial DNA variation differentially associates with the methylation status of articular cartilage by acting on key mechanisms involved in osteoarthritis, such as apoptosis and metabolic and developmental processes.
Collapse
Affiliation(s)
- Estefanía Cortés-Pereira
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | | | | | - María E Vázquez-Mosquera
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Sara Relaño
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Paula Ramos-Louro
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Alejandro Durán-Sotuela
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Andrea Dalmao-Fernández
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Natividad Oreiro
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Francisco J Blanco
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| | - Ignacio Rego-Pérez
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), and Universidade da Coruña, A Coruña, Spain
| |
Collapse
|
8
|
Marinello M, Werner A, Giannone M, Tahiri K, Alves S, Tesson C, den Dunnen W, Seeler JS, Brice A, Sittler A. SUMOylation by SUMO2 is implicated in the degradation of misfolded ataxin-7 via RNF4 in SCA7 models. Dis Model Mech 2019; 12:dmm.036145. [PMID: 30559154 PMCID: PMC6361149 DOI: 10.1242/dmm.036145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 12/04/2018] [Indexed: 01/10/2023] Open
Abstract
Perturbation of protein homeostasis and aggregation of misfolded proteins is a major cause of many human diseases. A hallmark of the neurodegenerative disease spinocerebellar ataxia type 7 (SCA7) is the intranuclear accumulation of mutant, misfolded ataxin-7 (polyQ-ATXN7). Here, we show that endogenous ATXN7 is modified by SUMO proteins, thus also suggesting a physiological role for this modification under conditions of proteotoxic stress caused by the accumulation of polyQ-ATXN7. Co-immunoprecipitation experiments, immunofluorescence microscopy and proximity ligation assays confirmed the colocalization and interaction of polyQ-ATXN7 with SUMO2 in cells. Moreover, upon inhibition of the proteasome, both endogenous SUMO2/3 and the RNF4 ubiquitin ligase surround large polyQ-ATXN7 intranuclear inclusions. Overexpression of RNF4 and/or SUMO2 significantly decreased levels of polyQ-ATXN7 and, upon proteasomal inhibition, led to a marked increase in the polyubiquitination of polyQ-ATXN7. This provides a mechanism for the clearance of polyQ-ATXN7 from affected cells that involves the recruitment of RNF4 by SUMO2/3-modified polyQ-ATXN7, thus leading to its ubiquitination and proteasomal degradation. In a SCA7 knock-in mouse model, we similarly observed colocalization of SUMO2/3 with polyQ-ATXN7 inclusions in the cerebellum and retina. Furthermore, we detected accumulation of SUMO2/3 high-molecular-mass species in the cerebellum of SCA7 knock-in mice, compared with their wild-type littermates, and changes in SUMO-related transcripts. Immunohistochemical analysis showed the accumulation of SUMO proteins and RNF4 in the cerebellum of SCA7 patients. Taken together, our results show that the SUMO pathway contributes to the clearance of aggregated ATXN7 and suggest that its deregulation might be associated with SCA7 disease progression.
Collapse
Affiliation(s)
- Martina Marinello
- Sorbonne Universités, UPMC, Univ Paris 06 UMRS 1127, INSERM U 1127, CNRS UMR 7225, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013 Paris, France.,Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Neurogenetics Group, 75013 Paris, France
| | - Andreas Werner
- Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Mariagiovanna Giannone
- Sorbonne Universités, UPMC, Univ Paris 06 UMRS 1127, INSERM U 1127, CNRS UMR 7225, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013 Paris, France.,Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Neurogenetics Group, 75013 Paris, France
| | - Khadija Tahiri
- Sorbonne Universités, UPMC, Univ Paris 06 UMRS 1127, INSERM U 1127, CNRS UMR 7225, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Sandro Alves
- Sorbonne Universités, UPMC, Univ Paris 06 UMRS 1127, INSERM U 1127, CNRS UMR 7225, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013 Paris, France
| | - Christelle Tesson
- Sorbonne Universités, UPMC, Univ Paris 06 UMRS 1127, INSERM U 1127, CNRS UMR 7225, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013 Paris, France.,Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences et Lettres (PSL) Research University, Neurogenetics Group, 75013 Paris, France
| | - Wilfred den Dunnen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Jacob-S Seeler
- Nuclear Organization and Oncogenesis Unit, INSERM U.993, Department of Cell Biology and Infection, Institut Pasteur, F-75015 Paris, France
| | - Alexis Brice
- Sorbonne Universités, UPMC, Univ Paris 06 UMRS 1127, INSERM U 1127, CNRS UMR 7225, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013 Paris, France.,AP-HP, Genetic Department, Pitié-Salpêtrière University Hospital, F-75013 Paris, France
| | - Annie Sittler
- Sorbonne Universités, UPMC, Univ Paris 06 UMRS 1127, INSERM U 1127, CNRS UMR 7225, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013 Paris, France
| |
Collapse
|
9
|
Molecular Mechanisms and Cellular Pathways Implicated in Machado-Joseph Disease Pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1049:349-367. [PMID: 29427113 DOI: 10.1007/978-3-319-71779-1_18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Machado-Joseph disease (MJD) is a dominantly inherited disorder originally described in people of Portuguese descent, and associated with the expansion of a CAG tract in the coding region of the causative gene MJD1/ATX3. The CAG repeats range from 10 to 51 in the normal population and from 55 to 87 in SCA3/MJD patients. MJD1 encodes ataxin-3, a protein whose physiological function has been linked to ubiquitin-mediated proteolysis. Despite the identification of the causative mutation, the pathogenic process leading to the neurodegeneration observed in the disease is not yet completely understood. In the past years, several studies identified different molecular mechanisms and cellular pathways as being impaired or deregulated in MJD. Autophagy, proteolysis or post-translational modifications, among other processes, were implicated in MJD pathogenesis. From these studies it was possible to identify new targets for therapeutic intervention, which in some cases proved successful in models of disease.
Collapse
|
10
|
The CAG-polyglutamine repeat diseases: a clinical, molecular, genetic, and pathophysiologic nosology. HANDBOOK OF CLINICAL NEUROLOGY 2018; 147:143-170. [PMID: 29325609 DOI: 10.1016/b978-0-444-63233-3.00011-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Throughout the genome, unstable tandem nucleotide repeats can expand to cause a variety of neurologic disorders. Expansion of a CAG triplet repeat within a coding exon gives rise to an elongated polyglutamine (polyQ) tract in the resultant protein product, and accounts for a unique category of neurodegenerative disorders, known as the CAG-polyglutamine repeat diseases. The nine members of the CAG-polyglutamine disease family include spinal and bulbar muscular atrophy (SBMA), Huntington disease, dentatorubral pallidoluysian atrophy, and six spinocerebellar ataxias (SCA 1, 2, 3, 6, 7, and 17). All CAG-polyglutamine diseases are dominantly inherited, with the exception of SBMA, which is X-linked, and many CAG-polyglutamine diseases display anticipation, which is defined as increasing disease severity in successive generations of an affected kindred. Despite widespread expression of the different polyQ-expanded disease proteins throughout the body, each CAG-polyglutamine disease strikes a particular subset of neurons, although the mechanism for this cell-type selectivity remains poorly understood. While the different genes implicated in these disorders display amino acid homology only in the repeat tract domain, certain pathologic molecular processes have been implicated in almost all of the CAG-polyglutamine repeat diseases, including protein aggregation, proteolytic cleavage, transcription dysregulation, autophagy impairment, and mitochondrial dysfunction. Here we highlight the clinical and molecular genetic features of each distinct disorder, and then discuss common themes in CAG-polyglutamine disease pathogenesis, closing with emerging advances in therapy development.
Collapse
|
11
|
Curtis HJ, Seow Y, Wood MJA, Varela MA. Knockdown and replacement therapy mediated by artificial mirtrons in spinocerebellar ataxia 7. Nucleic Acids Res 2017; 45:7870-7885. [PMID: 28575281 PMCID: PMC5569705 DOI: 10.1093/nar/gkx483] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 05/26/2017] [Indexed: 12/13/2022] Open
Abstract
We evaluate a knockdown-replacement strategy mediated by mirtrons as an alternative to allele-specific silencing using spinocerebellar ataxia 7 (SCA7) as a model. Mirtrons are introns that form pre-microRNA hairpins after splicing, producing RNAi effectors not processed by Drosha. Mirtron mimics may therefore avoid saturation of the canonical processing pathway. This method combines gene silencing mediated by an artificial mirtron with delivery of a functional copy of the gene such that both elements of the therapy are always expressed concurrently, minimizing the potential for undesirable effects and preserving wild-type function. This mutation- and single nucleotide polymorphism-independent method could be crucial in dominant diseases that feature both gain- and loss-of-function pathologies or have a heterogeneous genetic background. Here we develop mirtrons against ataxin 7 with silencing efficacy comparable to shRNAs, and introduce silent mutations into an ataxin 7 transgene such that it is resistant to their effect. We successfully express the transgene and one mirtron together from a single construct. Hence, we show that this method can be used to silence the endogenous allele of ataxin 7 and replace it with an exogenous copy of the gene, highlighting the efficacy and transferability across patient genotypes of this approach.
Collapse
Affiliation(s)
- Helen J Curtis
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK.,Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, UK
| | - Yiqi Seow
- Molecular Engineering Laboratory, Biomedical Sciences Institutes, A*STAR, Singapore
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Miguel A Varela
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| |
Collapse
|
12
|
Fernández‐Nogales M, Santos‐Galindo M, Merchán‐Rubira J, Hoozemans JJM, Rábano A, Ferrer I, Avila J, Hernández F, Lucas JJ. Tau-positive nuclear indentations in P301S tauopathy mice. Brain Pathol 2017; 27:314-322. [PMID: 27338164 PMCID: PMC8029483 DOI: 10.1111/bpa.12407] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/14/2016] [Indexed: 12/23/2022] Open
Abstract
Increased incidence of neuronal nuclear indentations is a well-known feature of the striatum of Huntington's disease (HD) brains and, in Alzheimer's disease (AD), neuronal nuclear indentations have recently been reported to correlate with neurotoxicity caused by improper cytoskeletal/nucleoskeletal coupling. Initial detection of rod-shaped tau immunostaining in nuclei of cortical and striatal neurons of HD brains and in hippocampal neurons of early Braak stage AD led us to coin the term "tau nuclear rods (TNRs)." Although TNRs traverse nuclear space, they in fact occupy narrow cytoplasmic extensions that fill indentations of the nuclear envelope and we will here refer to this histological hallmark as Tau-immunopositive nuclear indentations (TNIs). We reasoned that TNI formation is likely secondary to tau alterations as TNI detection in HD correlates with an increase in total tau, particularly of the isoforms with four tubulin binding repeats (4R-tau). Here we analyze transgenic mice that overexpress human 4R-tau with a frontotemporal lobar degeneration-tau point mutation (P301S mice) to explore whether tau alteration is sufficient for TNI formation. Immunohistochemistry with various tau antibodies, immunoelectron microscopy and double tau-immunofluorescence/DAPI-nuclear counterstaining confirmed that excess 4R-tau in P301S mice is sufficient for the detection of abundant TNIs that fill nuclear indentations. Interestingly, this does not correlate with an increase in the number of nuclear indentations, thus suggesting that excess total tau or an isoform imbalance in favor of 4R-tau facilitates tau detection inside preexisting nuclear indentations but does not induce formation of the latter. In summary, here we demonstrate that tau alteration is sufficient for TNI detection and our results suggest that the neuropathological finding of TNIs becomes a possible indicator of increased total tau and/or increased 4R/3R-tau ratio in the affected neurons apart from being an efficient way to monitor pathology-associated nuclear indentations.
Collapse
Affiliation(s)
- Marta Fernández‐Nogales
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAM28049MadridSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos IIISpain
| | - María Santos‐Galindo
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAM28049MadridSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos IIISpain
| | - Jesús Merchán‐Rubira
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAM28049MadridSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos IIISpain
| | - Jeroen J. M. Hoozemans
- Department of PathologyVU University Medical Center, Neuroscience Campus Amsterdam1007 MB Amsterdamthe Netherlands
| | - Alberto Rábano
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos IIISpain
- Departamento de Neuropatología y Banco de TejidosFundación CIENMadridSpain
| | - Isidro Ferrer
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos IIISpain
- Institute of Neuropathology; IDIBELL‐University Hospital BellvitgeUniversity of Barcelona; Hospitalet de LlobregatBarcelona08908Spain
| | - Jesús Avila
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAM28049MadridSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos IIISpain
| | - Félix Hernández
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAM28049MadridSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos IIISpain
| | - José J. Lucas
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAM28049MadridSpain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos IIISpain
| |
Collapse
|
13
|
Kuiper EFE, de Mattos EP, Jardim LB, Kampinga HH, Bergink S. Chaperones in Polyglutamine Aggregation: Beyond the Q-Stretch. Front Neurosci 2017; 11:145. [PMID: 28386214 PMCID: PMC5362620 DOI: 10.3389/fnins.2017.00145] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/08/2017] [Indexed: 01/12/2023] Open
Abstract
Expanded polyglutamine (polyQ) stretches in at least nine unrelated proteins lead to inherited neuronal dysfunction and degeneration. The expansion size in all diseases correlates with age at onset (AO) of disease and with polyQ protein aggregation, indicating that the expanded polyQ stretch is the main driving force for the disease onset. Interestingly, there is marked interpatient variability in expansion thresholds for a given disease. Between different polyQ diseases the repeat length vs. AO also indicates the existence of modulatory effects on aggregation of the upstream and downstream amino acid sequences flanking the Q expansion. This can be either due to intrinsic modulation of aggregation by the flanking regions, or due to differential interaction with other proteins, such as the components of the cellular protein quality control network. Indeed, several lines of evidence suggest that molecular chaperones have impact on the handling of different polyQ proteins. Here, we review factors differentially influencing polyQ aggregation: the Q-stretch itself, modulatory flanking sequences, interaction partners, cleavage of polyQ-containing proteins, and post-translational modifications, with a special focus on the role of molecular chaperones. By discussing typical examples of how these factors influence aggregation, we provide more insight on the variability of AO between different diseases as well as within the same polyQ disorder, on the molecular level.
Collapse
Affiliation(s)
- E F E Kuiper
- Department of Cell Biology, University Medical Center Groningen, University of Groningen Groningen, Netherlands
| | - Eduardo P de Mattos
- Department of Cell Biology, University Medical Center Groningen, University of GroningenGroningen, Netherlands; Programa de Pós-Graduação em Genética e Biologia Molecular, Department of Genetics, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto AlegrePorto Alegre, Brazil
| | - Laura B Jardim
- Programa de Pós-Graduação em Genética e Biologia Molecular, Department of Genetics, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil; Medical Genetics Service, Hospital de Clínicas de Porto AlegrePorto Alegre, Brazil; Departamento de Medicina Interna, Universidade Federal do Rio Grande do SulPorto Alegre, Brazil
| | - Harm H Kampinga
- Department of Cell Biology, University Medical Center Groningen, University of Groningen Groningen, Netherlands
| | - Steven Bergink
- Department of Cell Biology, University Medical Center Groningen, University of Groningen Groningen, Netherlands
| |
Collapse
|
14
|
Non-apoptotic cell death in animal development. Cell Death Differ 2017; 24:1326-1336. [PMID: 28211869 DOI: 10.1038/cdd.2017.20] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/18/2017] [Accepted: 01/19/2017] [Indexed: 01/22/2023] Open
Abstract
Programmed cell death (PCD) is an important process in the development of multicellular organisms. Apoptosis, a form of PCD characterized morphologically by chromatin condensation, membrane blebbing, and cytoplasm compaction, and molecularly by the activation of caspase proteases, has been extensively investigated. Studies in Caenorhabditis elegans, Drosophila, mice, and the developing chick have revealed, however, that developmental PCD also occurs through other mechanisms, morphologically and molecularly distinct from apoptosis. Some non-apoptotic PCD pathways, including those regulating germ cell death in Drosophila, still appear to employ caspases. However, another prominent cell death program, linker cell-type death (LCD), is morphologically conserved, and independent of the key genes that drive apoptosis, functioning, at least in part, through the ubiquitin proteasome system. These non-apoptotic processes may serve as backup programs when caspases are inactivated or unavailable, or, more likely, as freestanding cell culling programs. Non-apoptotic PCD has been documented extensively in the developing nervous system, and during the formation of germline and somatic gonadal structures, suggesting that preservation of these mechanisms is likely under strong selective pressure. Here, we discuss our current understanding of non-apoptotic PCD in animal development, and explore possible roles for LCD and other non-apoptotic developmental pathways in vertebrates. We raise the possibility that during vertebrate development, apoptosis may not be the major PCD mechanism.
Collapse
|
15
|
Fernández‐Nogales M, Santos‐Galindo M, Hernández IH, Cabrera JR, Lucas JJ. Faulty splicing and cytoskeleton abnormalities in Huntington's disease. Brain Pathol 2016; 26:772-778. [PMID: 27529534 PMCID: PMC8028924 DOI: 10.1111/bpa.12430] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 08/03/2016] [Indexed: 02/03/2023] Open
Abstract
Huntington's disease (HD) is caused by a CAG-repeat encoding a polyglutamine (polyQ) tract in the huntingtin protein. There is plenty of evidence of polyQ-driven toxicity. However, CAG repeat RNA-driven alteration of splicing has recently been proposed in analogy to CUG-repeat diseases. Here we review the reported alteration of the CAG-repeat associated splicing factor SRSF6 in brains of HD patients and mouse models and how this correlates with altered splicing of, at least, two microtubule-associated proteins in HD, namely MAPT (tau) and MAP2. Regarding tau, altered splicing of exon 10 has been reported, along with increased levels and 4R/3R-tau ratio and detection of tau in a new nuclear rod-shaped histopathological hallmark termed tau nuclear rod (TNR) or tau nuclear indentation (TNI). These findings, together with an attenuation of HD phenotype in R6/1 mice with tau deficiency and subsequent studies showing increased phosphorylation in mouse models and increased levels in CSF of patients, has led to proposing HD as a tauopathy. Regarding MAP2, an increase in its juvenile form and a decrease in total MAP2 together with redistribution from dendrites to soma is observed in HD patients, which may contribute to the dendritic atrophy in HD. Furthermore, MAP2 positive structures filling nuclear indentations have occasionally been found and co-localized with tau. Therefore, altered MAP function with imbalance in tau/MAP2 content could contribute to HD striatal atrophy and dysfunction. Besides, TNIs might be indicative of such MAP abnormalities. TNIs are also found in early pathology Alzheimer's disease and in tauopathy mice over-expressing mutant 4R-tau. This indicates that tau alteration is sufficient for TNI detection, which becomes a marker of increased total tau and/or altered 4R/3R-tau ratio and reporter of pathology-associated nuclear indentations. Altogether, these recent studies suggest that correcting the SRSF6-driven missplicing and/or microtubule-associated imbalance might be of therapeutic value in HD.
Collapse
Affiliation(s)
- Marta Fernández‐Nogales
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAMMadrid28049Spain
- Instituto de Salud Carlos IIINetworking Research Center on Neurodegenerative Diseases (CIBERNED)Spain
- Present address:
Present address: Marta Fernández‐Nogales, CSIC/University of Miguel HernándezInstituto De Neurociencias De Alicante (INA)AlicanteSpain
| | - María Santos‐Galindo
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAMMadrid28049Spain
- Instituto de Salud Carlos IIINetworking Research Center on Neurodegenerative Diseases (CIBERNED)Spain
| | - Ivó H. Hernández
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAMMadrid28049Spain
- Instituto de Salud Carlos IIINetworking Research Center on Neurodegenerative Diseases (CIBERNED)Spain
| | - Jorge R. Cabrera
- Department of Microbiology and ImmunologyDartmouth CollegeLebanonNH
| | - José J. Lucas
- Center for Molecular Biology “Severo Ochoa” (CBMSO) CSIC/UAMMadrid28049Spain
- Instituto de Salud Carlos IIINetworking Research Center on Neurodegenerative Diseases (CIBERNED)Spain
| |
Collapse
|
16
|
Schwendeman AR, Shaham S. A High-Throughput Small Molecule Screen for C. elegans Linker Cell Death Inhibitors. PLoS One 2016; 11:e0164595. [PMID: 27716809 PMCID: PMC5055323 DOI: 10.1371/journal.pone.0164595] [Citation(s) in RCA: 9] [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: 07/25/2016] [Accepted: 09/27/2016] [Indexed: 12/29/2022] Open
Abstract
Programmed cell death is a ubiquitous process in metazoan development. Apoptosis, one cell death form, has been studied extensively. However, mutations inactivating key mammalian apoptosis regulators do not block most developmental cell culling, suggesting that other cell death pathways are likely important. Recent work in the nematode Caenorhabditis elegans identified a non-apoptotic cell death form mediating the demise of the male-specific linker cell. This cell death process (LCD, linker cell-type death) is morphologically conserved, and its molecular effectors also mediate axon degeneration in mammals and Drosophila. To develop reagents to manipulate LCD, we established a simple high-throughput screening protocol for interrogating the effects of small molecules on C. elegans linker cell death in vivo. From 23,797 compounds assayed, 11 reproducibly block linker cell death onset. Of these, five induce animal lethality, and six promote a reversible developmental delay. These results provide proof-of principle validation of our screening protocol, demonstrate that developmental progression is required for linker cell death, and suggest that larger scale screens may identify LCD-specific small-molecule regulators that target the LCD execution machinery.
Collapse
Affiliation(s)
- Andrew R. Schwendeman
- Laboratory of Developmental Genetics, The Rockefeller University, New York, New York, United States of America
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
17
|
Transcriptome Profiling Identifies Multiplexin as a Target of SAGA Deubiquitinase Activity in Glia Required for Precise Axon Guidance During Drosophila Visual Development. G3-GENES GENOMES GENETICS 2016; 6:2435-45. [PMID: 27261002 PMCID: PMC4978897 DOI: 10.1534/g3.116.031310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Spt-Ada-Gcn5 Acetyltransferase (SAGA) complex is a transcriptional coactivator with histone acetylase and deubiquitinase activities that plays an important role in visual development and function. In Drosophila melanogaster, four SAGA subunits are required for the deubiquitination of monoubiquitinated histone H2B (ubH2B): Nonstop, Sgf11, E(y)2, and Ataxin 7. Mutations that disrupt SAGA deubiquitinase activity cause defects in neuronal connectivity in the developing Drosophila visual system. In addition, mutations in SAGA result in the human progressive visual disorder spinocerebellar ataxia type 7 (SCA7). Glial cells play a crucial role in both the neuronal connectivity defect in nonstop and sgf11 flies, and in the retinal degeneration observed in SCA7 patients. Thus, we sought to identify the gene targets of SAGA deubiquitinase activity in glia in the Drosophila larval central nervous system. To do this, we enriched glia from wild-type, nonstop, and sgf11 larval optic lobes using affinity-purification of KASH-GFP tagged nuclei, and then examined each transcriptome using RNA-seq. Our analysis showed that SAGA deubiquitinase activity is required for proper expression of 16% of actively transcribed genes in glia, especially genes involved in proteasome function, protein folding and axon guidance. We further show that the SAGA deubiquitinase-activated gene Multiplexin (Mp) is required in glia for proper photoreceptor axon targeting. Mutations in the human ortholog of Mp, COL18A1, have been identified in a family with a SCA7-like progressive visual disorder, suggesting that defects in the expression of this gene in SCA7 patients could play a role in the retinal degeneration that is unique to this ataxia.
Collapse
|
18
|
Alves S, Marais T, Biferi MG, Furling D, Marinello M, El Hachimi K, Cartier N, Ruberg M, Stevanin G, Brice A, Barkats M, Sittler A. Lentiviral vector-mediated overexpression of mutant ataxin-7 recapitulates SCA7 pathology and promotes accumulation of the FUS/TLS and MBNL1 RNA-binding proteins. Mol Neurodegener 2016; 11:58. [PMID: 27465358 PMCID: PMC4964261 DOI: 10.1186/s13024-016-0123-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 07/21/2016] [Indexed: 12/17/2022] Open
Abstract
Background We used lentiviral vectors (LVs) to generate a new SCA7 animal model overexpressing a truncated mutant ataxin-7 (MUT ATXN7) fragment in the mouse cerebellum, in order to characterize the specific neuropathological and behavioral consequences of the genetic defect in this brain structure. Results LV-mediated overexpression of MUT ATXN7 into the cerebellum of C57/BL6 adult mice induced neuropathological features similar to that observed in patients, such as intranuclear aggregates in Purkinje cells (PC), loss of synaptic markers, neuroinflammation, and neuronal death. No neuropathological changes were observed when truncated wild-type ataxin-7 (WT ATXN7) was injected. Interestingly, the local delivery of LV-expressing mutant ataxin-7 (LV-MUT-ATXN7) into the cerebellum of wild-type mice also mediated the development of an ataxic phenotype at 8 to 12 weeks post-injection. Importantly, our data revealed abnormal levels of the FUS/TLS, MBNL1, and TDP-43 RNA-binding proteins in the cerebellum of the LV-MUT-ATXN7 injected mice. MUT ATXN7 overexpression induced an increase in the levels of the pathological phosphorylated TDP-43, and a decrease in the levels of soluble FUS/TLS, with both proteins accumulating within ATXN7-positive intranuclear inclusions. MBNL1 also co-aggregated with MUT ATXN7 in most PC nuclear inclusions. Interestingly, no MBNL2 aggregation was observed in cerebellar MUT ATXN7 aggregates. Immunohistochemical studies in postmortem tissue from SCA7 patients and SCA7 knock-in mice confirmed SCA7-induced nuclear accumulation of FUS/TLS and MBNL1, strongly suggesting that these proteins play a physiopathological role in SCA7. Conclusions This study validates a novel SCA7 mouse model based on lentiviral vectors, in which strong and sustained expression of MUT ATXN7 in the cerebellum was found sufficient to generate motor defects. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0123-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sandro Alves
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités UPMC, Univ Paris 06 UMR_S 1127, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013, Paris, France.
| | - Thibaut Marais
- CNRS FRE3617, Center for Research in Myology, Sorbonne Universités UPMC Univ Paris 06, INSERM UMRS974, Institut de Myologie, G-H Pitié-Salpêtrière, 75013, Paris, France
| | - Maria-Grazia Biferi
- CNRS FRE3617, Center for Research in Myology, Sorbonne Universités UPMC Univ Paris 06, INSERM UMRS974, Institut de Myologie, G-H Pitié-Salpêtrière, 75013, Paris, France
| | - Denis Furling
- CNRS FRE3617, Center for Research in Myology, Sorbonne Universités UPMC Univ Paris 06, INSERM UMRS974, Institut de Myologie, G-H Pitié-Salpêtrière, 75013, Paris, France
| | - Martina Marinello
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités UPMC, Univ Paris 06 UMR_S 1127, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013, Paris, France.,EPHE Ecole Pratique des Hautes Etudes, Laboratoire de Neurogénétique, PSL Universités, 75013, Paris, France
| | - Khalid El Hachimi
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités UPMC, Univ Paris 06 UMR_S 1127, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013, Paris, France.,EPHE Ecole Pratique des Hautes Etudes, Laboratoire de Neurogénétique, PSL Universités, 75013, Paris, France
| | | | - Merle Ruberg
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités UPMC, Univ Paris 06 UMR_S 1127, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013, Paris, France
| | - Giovanni Stevanin
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités UPMC, Univ Paris 06 UMR_S 1127, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013, Paris, France.,EPHE Ecole Pratique des Hautes Etudes, Laboratoire de Neurogénétique, PSL Universités, 75013, Paris, France.,Département de Génétique et Cytogénétique, AP-HP, G-H Pitié-Salpêtrière, 47 Bd de l'Hôpital, 75013, Paris, France
| | - Alexis Brice
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités UPMC, Univ Paris 06 UMR_S 1127, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013, Paris, France.,Département de Génétique et Cytogénétique, AP-HP, G-H Pitié-Salpêtrière, 47 Bd de l'Hôpital, 75013, Paris, France
| | - Martine Barkats
- CNRS FRE3617, Center for Research in Myology, Sorbonne Universités UPMC Univ Paris 06, INSERM UMRS974, Institut de Myologie, G-H Pitié-Salpêtrière, 75013, Paris, France
| | - Annie Sittler
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités UPMC, Univ Paris 06 UMR_S 1127, ICM (Brain and Spine Institute) Pitié-Salpêtrière Hospital, 75013, Paris, France.
| |
Collapse
|
19
|
Yang H, Hu HY. Sequestration of cellular interacting partners by protein aggregates: implication in a loss-of-function pathology. FEBS J 2016; 283:3705-3717. [PMID: 27016044 DOI: 10.1111/febs.13722] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 03/11/2016] [Accepted: 03/24/2016] [Indexed: 01/09/2023]
Abstract
Protein misfolding and aggregation are a hallmark of several neurodegenerative diseases (NDs). However, how protein aggregation leads to cytotoxicity and neurodegeneration is still controversial. Emerging evidence demonstrates that sequestration of cellular-interacting partners by protein aggregates contributes to the pathogenesis of these diseases. Here, we review current research on sequestration of cellular proteins by protein aggregates and its relation to proteinopathies. Based on different interaction modes, we classify these protein sequestrations into four types: protein coaggregation, domain/motif-mediated sequestration, RNA-assisted sequestration, and sequestration of molecular chaperones. Thus, the cellular essential proteins and/or RNA hijacked by protein aggregates may lose their biological functions, consequently resulting in cytotoxicity and neurodegeneration. We have proposed a hijacking model recapitulating the sequestration process and the loss-of-function pathology of ND.
Collapse
Affiliation(s)
- Hui Yang
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hong-Yu Hu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
20
|
Cortes CJ, La Spada AR. Autophagy in polyglutamine disease: Imposing order on disorder or contributing to the chaos? Mol Cell Neurosci 2015; 66:53-61. [PMID: 25771431 DOI: 10.1016/j.mcn.2015.03.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/07/2015] [Accepted: 03/09/2015] [Indexed: 12/13/2022] Open
Abstract
Autophagy is an essential, fundamentally important catabolic pathway in which double membrane-bound vesicles form in the cytosol and encircle macromolecules and organelles to permit their degradation after fusion with lysosomes. More than a decade of research has revealed that autophagy is required for normal central nervous system (CNS) function and plays a central role in maintaining protein and organelle quality controls in neurons. Neurodegenerative diseases occur when misfolded proteins accumulate and disrupt normal cellular processes, and autophagy has emerged as a key arbiter of the cell's homeostatic response to this threat. One class of inherited neurodegenerative disease is known as the CAG/polyglutamine repeat disorders, and these diseases all result from the expansion of a CAG repeat tract in the coding regions of distinct genes. Polyglutamine (polyQ) repeat diseases result in the production polyQ-expanded proteins that misfold to form inclusions or aggregates that challenge the main cellular proteostasis system of the cell, the ubiquitin proteasome system (UPS). The UPS cannot efficiently degrade polyQ-expanded disease proteins, and components of the UPS are enriched in polyQ disease aggregate bodies found in degenerating neurons. In addition to components of the UPS, polyQ protein cytosolic aggregates co-localize with key autophagy proteins, even in autophagy deficient cells, suggesting that they probably do not reflect the formation of autophagosomes but rather the sequestration of key autophagy components. Furthermore, recent evidence now implicates polyQ proteins in the regulation of the autophagy pathway itself. Thus, a complex model emerges where polyQ proteins play a dual role as both autophagy substrates and autophagy offenders. In this review, we consider the role of autophagy in polyQ disorders and the therapeutic potential for autophagy modulation in these diseases. This article is part of a Special Issue entitled "Neuronal Protein".
Collapse
Affiliation(s)
- Constanza J Cortes
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92037, USA
| | - Albert R La Spada
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92037, USA; Department of Cellular & Molecular Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Neurosciences, University of California, San Diego, La Jolla, CA 92037, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Rady Children's Hospital, San Diego, CA 92193, USA.
| |
Collapse
|
21
|
Tan JY, Vance KW, Varela MA, Sirey T, Watson LM, Curtis HJ, Marinello M, Alves S, Steinkraus B, Cooper S, Nesterova T, Brockdorff N, Fulga T, Brice A, Sittler A, Oliver PL, Wood MJ, Ponting CP, Marques AC. Cross-talking noncoding RNAs contribute to cell-specific neurodegeneration in SCA7. Nat Struct Mol Biol 2014; 21:955-961. [PMID: 25306109 PMCID: PMC4255225 DOI: 10.1038/nsmb.2902] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 09/16/2014] [Indexed: 01/14/2023]
Abstract
What causes the tissue-specific pathology of diseases resulting from mutations in housekeeping genes? Specifically, in spinocerebellar ataxia type 7 (SCA7), a neurodegenerative disorder caused by a CAG-repeat expansion in ATXN7 (which encodes an essential component of the mammalian transcription coactivation complex, STAGA), the factors underlying the characteristic progressive cerebellar and retinal degeneration in patients were unknown. We found that STAGA is required for the transcription initiation of miR-124, which in turn mediates the post-transcriptional cross-talk between lnc-SCA7, a conserved long noncoding RNA, and ATXN7 mRNA. In SCA7, mutations in ATXN7 disrupt these regulatory interactions and result in a neuron-specific increase in ATXN7 expression. Strikingly, in mice this increase is most prominent in the SCA7 disease-relevant tissues, namely the retina and cerebellum. Our results illustrate how noncoding RNA-mediated feedback regulation of a ubiquitously expressed housekeeping gene may contribute to specific neurodegeneration.
Collapse
Affiliation(s)
- Jennifer Y Tan
- MRC Functional Genomics Unit, University of Oxford, Oxford, UK
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Keith W Vance
- MRC Functional Genomics Unit, University of Oxford, Oxford, UK
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Miguel A Varela
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Tamara Sirey
- MRC Functional Genomics Unit, University of Oxford, Oxford, UK
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Lauren M Watson
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
- University of Cape Town, Division of Human Genetics, Cape Town, South Africa
| | - Helen J Curtis
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Martina Marinello
- Centre de Recherche de l'Institut du Cerveau et de la Moëlle épinière, Hôpital de la Pitié-Salpêtrière, Paris, France
- Université Pierre et Marie Curie-Paris 6, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
- Département de Génétique et Cytogénétique, APHP, GH Pitié-Salpêtrière, Paris, France
| | - Sandro Alves
- Centre de Recherche de l'Institut du Cerveau et de la Moëlle épinière, Hôpital de la Pitié-Salpêtrière, Paris, France
- Université Pierre et Marie Curie-Paris 6, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
- Département de Génétique et Cytogénétique, APHP, GH Pitié-Salpêtrière, Paris, France
| | - Bruno Steinkraus
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Sarah Cooper
- University of Oxford, Department of Biochemistry, Oxford, United Kingdom
| | - Tatyana Nesterova
- University of Oxford, Department of Biochemistry, Oxford, United Kingdom
| | - Neil Brockdorff
- University of Oxford, Department of Biochemistry, Oxford, United Kingdom
| | - Tudor Fulga
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Alexis Brice
- Centre de Recherche de l'Institut du Cerveau et de la Moëlle épinière, Hôpital de la Pitié-Salpêtrière, Paris, France
- Université Pierre et Marie Curie-Paris 6, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
- Département de Génétique et Cytogénétique, APHP, GH Pitié-Salpêtrière, Paris, France
| | - Annie Sittler
- Centre de Recherche de l'Institut du Cerveau et de la Moëlle épinière, Hôpital de la Pitié-Salpêtrière, Paris, France
- Université Pierre et Marie Curie-Paris 6, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
| | - Peter L Oliver
- MRC Functional Genomics Unit, University of Oxford, Oxford, UK
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Matthew J Wood
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
- University of Cape Town, Division of Human Genetics, Cape Town, South Africa
| | - Chris P Ponting
- MRC Functional Genomics Unit, University of Oxford, Oxford, UK
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| | - Ana C Marques
- MRC Functional Genomics Unit, University of Oxford, Oxford, UK
- University of Oxford, Department of Physiology, Anatomy and Genetics, Oxford, United Kingdom
| |
Collapse
|
22
|
From pathways to targets: understanding the mechanisms behind polyglutamine disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:701758. [PMID: 25309920 PMCID: PMC4189765 DOI: 10.1155/2014/701758] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/03/2014] [Indexed: 12/27/2022]
Abstract
The history of polyglutamine diseases dates back approximately 20 years to the discovery of a polyglutamine repeat in the androgen receptor of SBMA followed by the identification of similar expansion mutations in Huntington's disease, SCA1, DRPLA, and the other spinocerebellar ataxias. This common molecular feature of polyglutamine diseases suggests shared mechanisms in disease pathology and neurodegeneration of disease specific brain regions. In this review, we discuss the main pathogenic pathways including proteolytic processing, nuclear shuttling and aggregation, mitochondrial dysfunction, and clearance of misfolded polyglutamine proteins and point out possible targets for treatment.
Collapse
|
23
|
Huntington's disease is a four-repeat tauopathy with tau nuclear rods. Nat Med 2014; 20:881-5. [PMID: 25038828 DOI: 10.1038/nm.3617] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 06/03/2014] [Indexed: 11/09/2022]
Abstract
An imbalance of tau isoforms containing either three or four microtubule-binding repeats causes frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17) in families with intronic mutations in the MAPT gene. Here we report equivalent imbalances at the mRNA and protein levels and increased total tau levels in the brains of subjects with Huntington's disease (HD) together with rod-like tau deposits along neuronal nuclei. These tau nuclear rods show an ordered filamentous ultrastructure and can be found filling the neuronal nuclear indentations previously reported in HD brains. Finally, alterations in serine/arginine-rich splicing factor-6 coincide with tau missplicing, and a role of tau in HD pathogenesis is evidenced by the attenuation of motor abnormalities of mutant HTT transgenic mice in tau knockout backgrounds.
Collapse
|
24
|
Tezenas du Montcel S, Durr A, Bauer P, Figueroa KP, Ichikawa Y, Brussino A, Forlani S, Rakowicz M, Schöls L, Mariotti C, van de Warrenburg BPC, Orsi L, Giunti P, Filla A, Szymanski S, Klockgether T, Berciano J, Pandolfo M, Boesch S, Melegh B, Timmann D, Mandich P, Camuzat A, Goto J, Ashizawa T, Cazeneuve C, Tsuji S, Pulst SM, Brusco A, Riess O, Brice A, Stevanin G. Modulation of the age at onset in spinocerebellar ataxia by CAG tracts in various genes. ACTA ACUST UNITED AC 2014; 137:2444-55. [PMID: 24972706 DOI: 10.1093/brain/awu174] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Polyglutamine-coding (CAG)n repeat expansions in seven different genes cause spinocerebellar ataxias. Although the size of the expansion is negatively correlated with age at onset, it accounts for only 50-70% of its variability. To find other factors involved in this variability, we performed a regression analysis in 1255 affected individuals with identified expansions (spinocerebellar ataxia types 1, 2, 3, 6 and 7), recruited through the European Consortium on Spinocerebellar Ataxias, to determine whether age at onset is influenced by the size of the normal allele in eight causal (CAG)n-containing genes (ATXN1-3, 6-7, 17, ATN1 and HTT). We confirmed the negative effect of the expanded allele and detected threshold effects reflected by a quadratic association between age at onset and CAG size in spinocerebellar ataxia types 1, 3 and 6. We also evidenced an interaction between the expanded and normal alleles in trans in individuals with spinocerebellar ataxia types 1, 6 and 7. Except for individuals with spinocerebellar ataxia type 1, age at onset was also influenced by other (CAG)n-containing genes: ATXN7 in spinocerebellar ataxia type 2; ATXN2, ATN1 and HTT in spinocerebellar ataxia type 3; ATXN1 and ATXN3 in spinocerebellar ataxia type 6; and ATXN3 and TBP in spinocerebellar ataxia type 7. This suggests that there are biological relationships among these genes. The results were partially replicated in four independent populations representing 460 Caucasians and 216 Asian samples; the differences are possibly explained by ethnic or geographical differences. As the variability in age at onset is not completely explained by the effects of the causative and modifier sister genes, other genetic or environmental factors must also play a role in these diseases.
Collapse
Affiliation(s)
- Sophie Tezenas du Montcel
- 1 Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, F-75013, Paris, France2 INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, F-75013, Paris, France3 AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Biostatistics Unit, Paris, F-75013, France
| | - Alexandra Durr
- 4 AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Genetics and Cytogenetics, F-75013, Paris, France5 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Peter Bauer
- 6 Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Karla P Figueroa
- 7 Department of Neurology, University of Utah, Salt Lake City, USA
| | - Yaeko Ichikawa
- 8 Department of Neurology, University of Tokyo, Graduate School of Medicine, Tokyo, Japan
| | - Alessandro Brussino
- 9 University of Torino, Department of Medical Sciences, and Medical Genetics Unit, Az. Osp. 'Città della Salute e della Scienza', Torino, Italy
| | - Sylvie Forlani
- 5 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Maria Rakowicz
- 10 Institute of Psychiatry and Neurology Warsaw, Sobieskiego 9, 02-957 Warsaw, Poland
| | - Ludger Schöls
- 11 Department of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany12 German Centre of Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Caterina Mariotti
- 13 SOSD Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS, Istituto Neurologico 'Carlo Besta', Milan, Italy
| | - Bart P C van de Warrenburg
- 14 Department of Neurology, Donders Institute for Brain, Cognition, and Behaviour, Radbound University Medical Centre, Nijmegen, The Netherlands
| | - Laura Orsi
- 15 Neurologic Division I, Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza, Torino, Italy
| | - Paola Giunti
- 16 Institute of Neurology, Department of Molecular Neuroscience, UCL, Queen Square, London, UK
| | - Alessandro Filla
- 17 Department of Neurological Sciences, Federico II University, Naples, Italy
| | - Sandra Szymanski
- 18 Department of Neurology, St. Josef Hospital, University Hospital of Bochum, Bochum, Germany
| | | | - José Berciano
- 20 Department of Neurology, University Hospital 'Marqués de Valdecilla', UC, IDIVAL and CIBERNED, 39008 Santander, Spain
| | - Massimo Pandolfo
- 21 Department of Neurology, ULB-Hôpital Erasme, Université Libre de Bruxelles, CP 231, Campus Plaine, ULB, Brusssels, Belgium
| | - Sylvia Boesch
- 22 Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Bela Melegh
- 23 Department of Medical Genetics, and Szentagothai Research Centre, University Pécs, Hungary
| | - Dagmar Timmann
- 24 Department of Neurology, University Clinic Essen, University of Duisburg-Essen, Essen, Germany
| | - Paola Mandich
- 25 Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal Child Health, University of Genova, and U.O. Medical Genetics of IRCCS AOU S. Martino Institute, Genova, Italy
| | - Agnès Camuzat
- 5 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | | | | | - Jun Goto
- 8 Department of Neurology, University of Tokyo, Graduate School of Medicine, Tokyo, Japan
| | - Tetsuo Ashizawa
- 26 Department of Neurology and McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Cécile Cazeneuve
- 4 AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Genetics and Cytogenetics, F-75013, Paris, France
| | - Shoji Tsuji
- 8 Department of Neurology, University of Tokyo, Graduate School of Medicine, Tokyo, Japan
| | - Stefan-M Pulst
- 7 Department of Neurology, University of Utah, Salt Lake City, USA
| | - Alfredo Brusco
- 9 University of Torino, Department of Medical Sciences, and Medical Genetics Unit, Az. Osp. 'Città della Salute e della Scienza', Torino, Italy
| | - Olaf Riess
- 6 Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Alexis Brice
- 4 AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Genetics and Cytogenetics, F-75013, Paris, France5 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Giovanni Stevanin
- 4 AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Genetics and Cytogenetics, F-75013, Paris, France5 Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France27 Ecole Pratique des Hautes Etudes, heSam Université, laboratoire de neurogénétique, ICM, Groupe Hospitalier Pitié-Salpêtrière, F-75013 Paris, France
| |
Collapse
|
25
|
Matilla-Dueñas A, Ashizawa T, Brice A, Magri S, McFarland KN, Pandolfo M, Pulst SM, Riess O, Rubinsztein DC, Schmidt J, Schmidt T, Scoles DR, Stevanin G, Taroni F, Underwood BR, Sánchez I. Consensus paper: pathological mechanisms underlying neurodegeneration in spinocerebellar ataxias. CEREBELLUM (LONDON, ENGLAND) 2014; 13:269-302. [PMID: 24307138 PMCID: PMC3943639 DOI: 10.1007/s12311-013-0539-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intensive scientific research devoted in the recent years to understand the molecular mechanisms or neurodegeneration in spinocerebellar ataxias (SCAs) are identifying new pathways and targets providing new insights and a better understanding of the molecular pathogenesis in these diseases. In this consensus manuscript, the authors discuss their current views on the identified molecular processes causing or modulating the neurodegenerative phenotype in spinocerebellar ataxias with the common opinion of translating the new knowledge acquired into candidate targets for therapy. The following topics are discussed: transcription dysregulation, protein aggregation, autophagy, ion channels, the role of mitochondria, RNA toxicity, modulators of neurodegeneration and current therapeutic approaches. Overall point of consensus includes the common vision of neurodegeneration in SCAs as a multifactorial, progressive and reversible process, at least in early stages. Specific points of consensus include the role of the dysregulation of protein folding, transcription, bioenergetics, calcium handling and eventual cell death with apoptotic features of neurons during SCA disease progression. Unresolved questions include how the dysregulation of these pathways triggers the onset of symptoms and mediates disease progression since this understanding may allow effective treatments of SCAs within the window of reversibility to prevent early neuronal damage. Common opinions also include the need for clinical detection of early neuronal dysfunction, for more basic research to decipher the early neurodegenerative process in SCAs in order to give rise to new concepts for treatment strategies and for the translation of the results to preclinical studies and, thereafter, in clinical practice.
Collapse
Affiliation(s)
- A Matilla-Dueñas
- Health Sciences Research Institute Germans Trias i Pujol (IGTP), Ctra. de Can Ruti, Camí de les Escoles s/n, Badalona, Barcelona, Spain,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yu X, Muñoz-Alarcón A, Ajayi A, Webling KE, Steinhof A, Langel Ü, Ström AL. Inhibition of autophagy via p53-mediated disruption of ULK1 in a SCA7 polyglutamine disease model. J Mol Neurosci 2013; 50:586-99. [PMID: 23592174 DOI: 10.1007/s12031-013-0012-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 04/01/2013] [Indexed: 02/07/2023]
Abstract
Spinocerebellar ataxia type 7 (SCA7) is one of nine neurodegenerative disorders caused by expanded polyglutamine domains. These so-called polyglutamine (polyQ) diseases are all characterized by aggregation. Reducing the level of aggregating polyQ proteins via pharmacological activation of autophagy has been suggested as a therapeutic approach. However, recently, evidence implicating autophagic dysfunction in these disorders has also been reported. In this study, we show that the SCA7 polyglutamine protein ataxin-7 (ATXN7) reduces the autophagic activity via a previously unreported mechanism involving p53-mediated disruption of two key proteins involved in autophagy initiation. We show that in mutant ATXN7 cells, an increased p53-FIP200 interaction and co-aggregation of p53-FIP200 into ATXN7 aggregates result in decreased soluble FIP200 levels and subsequent destabilization of ULK1. Together, this leads to a decreased capacity for autophagy induction via the ULK1-FIP200-Atg13-Atg101 complex. We also show that treatment with a p53 inhibitor, or a blocker of ATXN7 aggregation, can restore the soluble levels of FIP200 and ULK1, as well as increase the autophagic activity and reduce ATXN7 toxicity. Understanding the mechanism behind polyQ-mediated inhibition of autophagy is of importance if therapeutic approaches based on autophagy stimulation should be developed for these disorders.
Collapse
Affiliation(s)
- Xin Yu
- Department of Neurochemistry, Stockholm University, 106 91, Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
27
|
Chort A, Alves S, Marinello M, Dufresnois B, Dornbierer JG, Tesson C, Latouche M, Baker DP, Barkats M, El Hachimi KH, Ruberg M, Janer A, Stevanin G, Brice A, Sittler A. Interferon beta induces clearance of mutant ataxin 7 and improves locomotion in SCA7 knock-in mice. Brain 2013; 136:1732-45. [DOI: 10.1093/brain/awt061] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
28
|
Blum ES, Schwendeman AR, Shaham S. PolyQ disease: misfiring of a developmental cell death program? Trends Cell Biol 2012; 23:168-74. [PMID: 23228508 DOI: 10.1016/j.tcb.2012.11.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 11/06/2012] [Accepted: 11/12/2012] [Indexed: 12/14/2022]
Abstract
Polyglutamine (polyQ) repeat diseases are neurodegenerative ailments elicited by glutamine-encoding CAG nucleotide expansions within endogenous human genes. Despite efforts to understand the basis of these diseases, the precise mechanism of cell death remains stubbornly unclear. Much of the data seem to be consistent with a model in which toxicity is an inherent property of the polyQ repeat, whereas host protein sequences surrounding the polyQ expansion modulate severity, age of onset, and cell specificity. Recently, a gene, pqn-41, encoding a glutamine-rich protein, was found to promote normally occurring non-apoptotic cell death in Caenorhabditis elegans. Here we review evidence for toxic and modulatory roles for polyQ repeats and their host proteins, respectively, and suggest similarities with pqn-41 function. We explore the hypothesis that toxicity mediated by glutamine-rich motifs may be important not only in pathology, but also in normal development.
Collapse
Affiliation(s)
- Elyse S Blum
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | | | | |
Collapse
|
29
|
Spinocerebellar ataxia type 35 (SCA35)-associated transglutaminase 6 mutants sensitize cells to apoptosis. Biochem Biophys Res Commun 2012. [PMID: 23206699 DOI: 10.1016/j.bbrc.2012.11.069] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Spinocerebellar ataxia type 35 (SCA35) is an autosomal dominant neurodegenerative disorder. In our previous study, using exome sequencing and linkage analysis, two missense mutations of the transglutaminase 6 (TGM6) gene were identified as causative for SCA35. TGM6 encodes transglutaminase 6 (TG6), a member of the transglutaminase family of enzymes that catalyze the formation of a covalent bond between a free amine group and the γ-carboxamide group of protein- or peptide-bound glutamine. However, the precise role of TG6 in contributing to SCA35 remains unclear. In this study, we analyzed the subcellular distribution, expression and in vitro activity of two missense mutations of TG6 (D327G, L517W) and found that both mutants exhibited decreased transglutaminase activity and stability. Furthermore, overexpressing the TG6 mutants sensitized cells to staurosporine-induced apoptosis by increasing the activity of caspases. We propose that the pro-apoptotic role of these mutants might underlie the pathogenesis of SCA35.
Collapse
|
30
|
Yanicostas C, Barbieri E, Hibi M, Brice A, Stevanin G, Soussi-Yanicostas N. Requirement for zebrafish ataxin-7 in differentiation of photoreceptors and cerebellar neurons. PLoS One 2012; 7:e50705. [PMID: 23226359 PMCID: PMC3511343 DOI: 10.1371/journal.pone.0050705] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 10/24/2012] [Indexed: 11/25/2022] Open
Abstract
The expansion of a polyglutamine (polyQ) tract in the N-terminal region of ataxin-7 (atxn7) is the causative event in spinocerebellar ataxia type 7 (SCA7), an autosomal dominant neurodegenerative disorder mainly characterized by progressive, selective loss of rod-cone photoreceptors and cerebellar Purkinje and granule cells. The molecular and cellular processes underlying this restricted neuronal vulnerability, which contrasts with the broad expression pattern of atxn7, remains one of the most enigmatic features of SCA7, and more generally of all polyQ disorders. To gain insight into this specific neuronal vulnerability and achieve a better understanding of atxn7 function, we carried out a functional analysis of this protein in the teleost fish Danio rerio. We characterized the zebrafish atxn7 gene and its transcription pattern, and by making use of morpholino-oligonucleotide-mediated gene inactivation, we analysed the phenotypes induced following mild or severe zebrafish atxn7 depletion. Severe or nearly complete zebrafish atxn7 loss-of-function markedly impaired embryonic development, leading to both early embryonic lethality and severely deformed embryos. More importantly, in relation to SCA7, moderate depletion of the protein specifically, albeit partially, prevented the differentiation of both retina photoreceptors and cerebellar Purkinje and granule cells. In addition, [1–232] human atxn7 fragment rescued these phenotypes showing strong function conservation of this protein through evolution. The specific requirement for zebrafish atxn7 in the proper differentiation of cerebellar neurons provides, to our knowledge, the first in vivo evidence of a direct functional relationship between atxn7 and the differentiation of Purkinje and granule cells, the most crucial neurons affected in SCA7 and most other polyQ-mediated SCAs. These findings further suggest that altered protein function may play a role in the pathophysiology of the disease, an important step toward the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Constantin Yanicostas
- INSERM, U676, Hôpital Robert Debré, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Elisa Barbieri
- INSERM, U676, Hôpital Robert Debré, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- INSERM, U975, Paris, France
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, UMR_S975, GHU Pitié-Salpêtrière, Paris, France
- CNRS, UMR7225, Paris, France
| | - Masahiko Hibi
- Laboratory for Vertebrate Axis Formation, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
- Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan
| | - Alexis Brice
- INSERM, U975, Paris, France
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, UMR_S975, GHU Pitié-Salpêtrière, Paris, France
- CNRS, UMR7225, Paris, France
| | - Giovanni Stevanin
- INSERM, U975, Paris, France
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, UMR_S975, GHU Pitié-Salpêtrière, Paris, France
- CNRS, UMR7225, Paris, France
- Ecole Pratique des Hautes Etudes, Paris, France
| | - Nadia Soussi-Yanicostas
- INSERM, U676, Hôpital Robert Debré, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
31
|
Blum ES, Abraham MC, Yoshimura S, Lu Y, Shaham S. Control of nonapoptotic developmental cell death in Caenorhabditis elegans by a polyglutamine-repeat protein. Science 2012; 335:970-3. [PMID: 22363008 DOI: 10.1126/science.1215156] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Death is a vital developmental cell fate. In Caenorhabditis elegans, programmed death of the linker cell, which leads gonadal elongation, proceeds independently of caspases and apoptotic effectors. To identify genes promoting linker-cell death, we performed a genome-wide RNA interference screen. We show that linker-cell death requires the gene pqn-41, encoding an endogenous polyglutamine-repeat protein. pqn-41 functions cell-autonomously and is expressed at the onset of linker-cell death. pqn-41 expression is controlled by the mitogen-activated protein kinase kinase SEK-1, which functions in parallel to the zinc-finger protein LIN-29 to promote cellular demise. Linker-cell death is morphologically similar to cell death associated with normal vertebrate development and polyglutamine-induced neurodegeneration. Our results may therefore provide molecular inroads to understanding nonapoptotic cell death in metazoan development and disease.
Collapse
Affiliation(s)
- Elyse S Blum
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
Spinocerebellar ataxia type 7 (SCA7) is associated with progressive blindness, dominant transmission, and marked anticipation. SCA7 represents one of the polyglutamine expansion diseases with increase of CAG repeats. The gene maps to chromosome 3p12-p21.1. Normal values of CAG repeats range from 4 to 18. The SCA7 gene encodes a protein of largely unknown function, called ataxin-7. SCA7 is reported in many countries and ethnic groups. Its phenotypic expression depends on the number of expanded repeats. The infantile phenotype is very severe, with more than 100 repeats. The classic type has 50 to 55 repeats and is characterized by a combination of visual and ataxic disturbances lasting for 20-40 years.When the number of CAG repeats is between 36 and 43, the evolution is much slower, with few or no retinal abnormalities. A CAG repeat number from 18 to 35 is asymptomatic but predisposes to the development of the disorder when expanding to the pathological range through transmission. The diagnosis is made by molecular genetics. The neuropathology of the disorder includes atrophy of the spinocerebellar pathways, pyramidal tracts, and motor nuclei in the brainstem and spinal cord, a cone-rod sytrophy of the retina, and ataxin-7 immunoreactive neuronal intranuclear inclusions. The neuropathological features vary as a function of the number of CAG repeats. Present research deals mainly with the study of ataxin-7 in transfected neural cells and transgenic mouse models.
Collapse
|
33
|
Matos CA, de Macedo-Ribeiro S, Carvalho AL. Polyglutamine diseases: The special case of ataxin-3 and Machado–Joseph disease. Prog Neurobiol 2011; 95:26-48. [DOI: 10.1016/j.pneurobio.2011.06.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 06/06/2011] [Accepted: 06/07/2011] [Indexed: 10/18/2022]
|
34
|
Mallik M, Lakhotia SC. Modifiers and mechanisms of multi-system polyglutamine neurodegenerative disorders: lessons from fly models. J Genet 2010; 89:497-526. [DOI: 10.1007/s12041-010-0072-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
35
|
Hsp105 reduces the protein aggregation and cytotoxicity by expanded-polyglutamine proteins through the induction of Hsp70. Exp Cell Res 2010; 316:2424-33. [DOI: 10.1016/j.yexcr.2010.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 06/03/2010] [Accepted: 06/06/2010] [Indexed: 11/20/2022]
|
36
|
La Spada AR, Taylor JP. Repeat expansion disease: progress and puzzles in disease pathogenesis. Nat Rev Genet 2010; 11:247-58. [PMID: 20177426 PMCID: PMC4704680 DOI: 10.1038/nrg2748] [Citation(s) in RCA: 336] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Repeat expansion mutations cause at least 22 inherited neurological diseases. The complexity of repeat disease genetics and pathobiology has revealed unexpected shared themes and mechanistic pathways among the diseases, such as RNA toxicity. Also, investigation of the polyglutamine diseases has identified post-translational modification as a key step in the pathogenic cascade and has shown that the autophagy pathway has an important role in the degradation of misfolded proteins--two themes that are likely to be relevant to the entire neurodegeneration field. Insights from repeat disease research are catalysing new lines of study that should not only elucidate molecular mechanisms of disease but also highlight opportunities for therapeutic intervention for these currently untreatable disorders.
Collapse
Affiliation(s)
- Albert R La Spada
- Division of Genetics, Department of Pediatrics, Institute for Genomic Medicine, University of California-San Diego, La Jolla, California 92093, USA.
| | | |
Collapse
|
37
|
Janer A, Werner A, Takahashi-Fujigasaki J, Daret A, Fujigasaki H, Takada K, Duyckaerts C, Brice A, Dejean A, Sittler A. SUMOylation attenuates the aggregation propensity and cellular toxicity of the polyglutamine expanded ataxin-7. Hum Mol Genet 2010; 19:181-95. [PMID: 19843541 DOI: 10.1093/hmg/ddp478] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Post-translational modification by SUMO (small ubiquitin-like modifier) was proposed to modulate the pathogenesis of several neurodegenerative diseases. Spinocerebellar ataxia type 7 (SCA7) is a neurodegenerative disorder, whose pathology is caused by an expansion of a polyglutamine stretch in the protein ataxin-7 (ATXN7). Here, we identified ATXN7 as new target for SUMOylation in vitro and in vivo. The major SUMO acceptor site was mapped to lysine 257, which is part of an evolutionarily conserved consensus SUMOylation motif. SUMOylation did not influence the subcellular localization of ATXN7 nor its interaction with components of the TFTC/STAGA complex. Expansion of the polyglutamine stretch did not impair the SUMOylation of ATXN7. Furthermore, SUMO1 and SUMO2 colocalized with ATXN7 in a subset of neuronal intranuclear inclusions in the brain of SCA7 patients and SCA7 knock-in mice. In a COS-7 cellular model of SCA7, in addition to diffuse nucleoplasmic staining we identified two populations of nuclear inclusions: homogenous or non-homogenous. Non-homogenous inclusions showed significantly reduced colocalization with SUMO1 and SUMO2, but were highly enriched in Hsp70, 19S proteasome and ubiquitin. Interestingly, they were characterized by increased staining with the apoptotic marker caspase-3 and by disruption of PML nuclear bodies. Importantly, preventing the SUMOylation of expanded ATXN7 by mutating the SUMO site increased both the amount of SDS-insoluble aggregates and of caspase-3 positive non-homogenous inclusions, which act toxic to the cells. Our results demonstrate an influence of SUMOylation on the multistep aggregation process of ATXN7 and implicate a role for ATXN7 SUMOylation in SCA7 pathogenesis.
Collapse
|
38
|
Polyglutamine-expanded ataxin-7 causes cerebellar dysfunction by inducing transcriptional dysregulation. Neurochem Int 2010; 56:329-39. [DOI: 10.1016/j.neuint.2009.11.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 11/04/2009] [Accepted: 11/04/2009] [Indexed: 11/23/2022]
|
39
|
Posttranslational modification of ataxin-7 at lysine 257 prevents autophagy-mediated turnover of an N-terminal caspase-7 cleavage fragment. J Neurosci 2009; 29:15134-44. [PMID: 19955365 DOI: 10.1523/jneurosci.4720-09.2009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Polyglutamine (polyQ) expansion within the ataxin-7 protein, a member of the STAGA [SPT3-TAF(II)31-GCN5L acetylase] and TFTC (GCN5 and TRRAP) chromatin remodeling complexes, causes the neurodegenerative disease spinocerebellar ataxia type 7 (SCA7). Proteolytic processing of ataxin-7 by caspase-7 generates N-terminal toxic polyQ-containing fragments that accumulate with disease progression and play an important role in SCA7 pathogenesis. To elucidate the basis for the toxicity of these fragments, we evaluated which posttranslational modifications of the N-terminal fragment of ataxin-7 modulate turnover and toxicity. Here, we show that mutating lysine 257 (K257), an amino acid adjacent to the caspase-7 cleavage site of ataxin-7 regulates turnover of the truncation product in a repeat-dependent manner. Modification of ataxin-7 K257 by acetylation promotes accumulation of the fragment, while unmodified ataxin-7 is degraded. The degradation of the caspase-7 cleavage product is mediated by macroautophagy in cell culture and primary neuron models of SCA7. Consistent with this, the fragment colocalizes with autophagic vesicle markers, and enhanced fragment accumulation increases in these lysosomal structures. We suggest that the levels of fragment accumulation within the cell is a key event in SCA7 neurodegeneration, and enhancing clearance of polyQ-containing fragments may be an effective target to reduce neurotoxicity in SCA7.
Collapse
|
40
|
Design of RNAi hairpins for mutation-specific silencing of ataxin-7 and correction of a SCA7 phenotype. PLoS One 2009; 4:e7232. [PMID: 19789634 PMCID: PMC2747278 DOI: 10.1371/journal.pone.0007232] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Accepted: 09/01/2009] [Indexed: 11/19/2022] Open
Abstract
Spinocerebellar ataxia type 7 is a polyglutamine disorder caused by an expanded CAG repeat mutation that results in neurodegeneration. Since no treatment exists for this chronic disease, novel therapies such post-transcriptional RNA interference-based gene silencing are under investigation, in particular those that might enable constitutive and tissue-specific silencing, such as expressed hairpins. Given that this method of silencing can be abolished by the presence of nucleotide mismatches against the target RNA, we sought to identify expressed RNA hairpins selective for silencing the mutant ataxin-7 transcript using a linked SNP. By targeting both short and full-length tagged ataxin-7 sequences, we show that mutation-specific selectivity can be obtained with single nucleotide mismatches to the wild-type RNA target incorporated 3′ to the centre of the active strand of short hairpin RNAs. The activity of the most effective short hairpin RNA incorporating the nucleotide mismatch at position 16 was further studied in a heterozygous ataxin-7 disease model, demonstrating significantly reduced levels of toxic mutant ataxin-7 protein with decreased mutant protein aggregation and retention of normal wild-type protein in a non-aggregated diffuse cellular distribution. Allele-specific mutant ataxin7 silencing was also obtained with the use of primary microRNA mimics, the most highly effective construct also harbouring the single nucleotide mismatch at position 16, corroborating our earlier findings. Our data provide understanding of RNA interference guide strand anatomy optimised for the allele-specific silencing of a polyglutamine mutation linked SNP and give a basis for the use of allele-specific RNA interference as a viable therapeutic approach for spinocerebellar ataxia 7.
Collapse
|
41
|
Bauer PO, Nukina N. The pathogenic mechanisms of polyglutamine diseases and current therapeutic strategies. J Neurochem 2009; 110:1737-65. [PMID: 19650870 DOI: 10.1111/j.1471-4159.2009.06302.x] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Expansion of CAG trinucleotide repeat within the coding region of several genes results in the production of proteins with expanded polyglutamine (PolyQ) stretch. The expression of these pathogenic proteins leads to PolyQ diseases, such as Huntington's disease or several types of spinocerebellar ataxias. This family of neurodegenerative disorders is characterized by constant progression of the symptoms and molecularly, by the accumulation of mutant proteins inside neurons causing their dysfunction and eventually death. So far, no effective therapy actually preventing the physical and/or mental decline has been developed. Experimental therapeutic strategies either target the levels or processing of mutant proteins in an attempt to prevent cellular deterioration, or they are aimed at the downstream pathologic effects to reverse or ameliorate the caused damages. Certain pathomechanistic aspects of PolyQ disorders are discussed here. Relevance of disease models and recent knowledge of therapeutic possibilities is reviewed and updated.
Collapse
Affiliation(s)
- Peter O Bauer
- Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Saitama, Japan
| | | |
Collapse
|
42
|
Fujinaga R, Takeshita Y, Uozumi K, Yanai A, Yoshioka K, Kokubu K, Shinoda K. Microtubule-dependent formation of the stigmoid body as a cytoplasmic inclusion distinct from pathological aggresomes. Histochem Cell Biol 2009; 132:305-18. [DOI: 10.1007/s00418-009-0618-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2009] [Indexed: 11/29/2022]
|
43
|
Fu L, Sztul E. ER-associated complexes (ERACs) containing aggregated cystic fibrosis transmembrane conductance regulator (CFTR) are degraded by autophagy. Eur J Cell Biol 2009; 88:215-26. [PMID: 19131141 DOI: 10.1016/j.ejcb.2008.11.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 11/20/2008] [Accepted: 11/21/2008] [Indexed: 11/18/2022] Open
Abstract
The ubiquitin-proteasome pathway and autophagy are the two major mechanisms responsible for the clearance of cellular proteins. We have used the yeast Saccharomyces cerevisiae as a model system and the cystic fibrosis transmembrane conductance regulator (CFTR) as a model substrate to study the interactive function of these two pathways in the degradation of misfolded proteins. EGFP-tagged human CFTR was introduced into yeast and expressed under a copper-inducible promoter. The localization and degradation of EGFP-CFTR in live cells were monitored by time-lapse imaging following its de novo synthesis. EGFP-CFTR first appears within the perinuclear and sub-cortical ER and is mobile within the plane of the membrane as assessed by fluorescence recovery after photobleaching (FRAP). This pool of EGFP-CFTR is subsequently degraded through a proteasome-dependent pathway that is inhibited in the pre1-1 yeast strain defective in proteasomal degradation. Prolonged expression of EGFP-CFTR leads to the sequestration of EGFP-CFTR molecules into ER structures called ER-associated complexes (ERACs). The sequestration of EGFP-CFTR into ERACs appears to be driven by aggregation since EGFP-CFTR molecules present within ERACs are immobile as measured by FRAP. Individual ERACs are cleared from cells through the autophagic pathway that is blocked in the atg6Delta and atg1Delta yeast strains defective in autophagy. Our results suggest that the proteasomal and the autophagic pathways function together to clear misfolded proteins from the ER.
Collapse
Affiliation(s)
- Lianwu Fu
- Department of Cell Biology, University of Alabama at Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA.
| | | |
Collapse
|
44
|
Kundu M, Thompson CB. Autophagy: basic principles and relevance to disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2008; 3:427-55. [PMID: 18039129 DOI: 10.1146/annurev.pathmechdis.2.010506.091842] [Citation(s) in RCA: 410] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Autophagy is a process by which cytoplasmic components are sequestered in double membrane vesicles and degraded upon fusion with lysosomal compartments. In yeast, autophagy is activated in response to changes in the extracellular milieu. Depending upon the stimulus, autophagy can degrade cytoplasmic contents nonspecifically or can target the degradation of specific cellular components. Both of these have been adopted in higher eukaryotes and account for the expanding role of autophagy in various cellular processes, as well as contribute to the variation in cellular outcomes after induction of autophagy. In some cases, autophagy appears to be an adaptive response, whereas under other circumstances it is involved in cell death. In mammals, autophagy has been implicated in either the pathogenesis or response to a wide variety of diseases, including neurodegenerative disease, chronic bacterial and viral infections, atherosclerosis, and cancer. As the basic molecular pathways that regulate autophagy are elucidated, the relationship of autophagy to the pathogenesis of various disease states emerges.
Collapse
Affiliation(s)
- Mondira Kundu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19063, USA.
| | | |
Collapse
|
45
|
Tarlac V, Turnbull V, Stefani D, Kelly L, Walsh R, Storey E. Inclusion formation by ataxins -1, -2, -3, and -7. Int J Neurosci 2007; 117:1289-314. [PMID: 17654093 DOI: 10.1080/00207450600936668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The authors studied inclusion formation in vitro using transiently transfected PC12 cells, with epitope-tagged and untagged full-length and truncated wild-type and expanded ataxins -1, -2, -3, and -7. At 72 hours, no inclusions were seen with wild-type full-length or truncated ataxins -2, -3, or -7, and only one with ataxin-1. Truncation abolished nuclear localization of ataxins -1 and -7, and allowed nuclear entry of ataxin-2. Of the expanded ataxins, only -1 and -2 formed inclusions, and those of ataxin-2 were rare and exclusively cytoplasmic. Truncation resulted in inclusion formation by ataxins -3 and -7, increased ataxin-1 inclusions, and enabled formation of nuclear ataxin-2 inclusions. There was no recruitment of wild-type ataxin-1 to expanded ataxin-1 inclusions.
Collapse
Affiliation(s)
- Volga Tarlac
- Van Cleef Roet Centre for Nervous Diseases, Department of Medicine (Neuroscience), Monash University (Alfred Hospital Campus), Melbourne, VIC, Australia
| | | | | | | | | | | |
Collapse
|
46
|
Young JE, Gouw L, Propp S, Sopher BL, Taylor J, Lin A, Hermel E, Logvinova A, Chen SF, Chen S, Bredesen DE, Truant R, Ptacek LJ, La Spada AR, Ellerby LM. Proteolytic cleavage of ataxin-7 by caspase-7 modulates cellular toxicity and transcriptional dysregulation. J Biol Chem 2007; 282:30150-60. [PMID: 17646170 DOI: 10.1074/jbc.m705265200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a polyglutamine (polyQ) disorder characterized by specific degeneration of cerebellar, brainstem, and retinal neurons. Although they share little sequence homology, proteins implicated in polyQ disorders have common properties beyond their characteristic polyQ tract. These include the production of proteolytic fragments, nuclear accumulation, and processing by caspases. Here we report that ataxin-7 is cleaved by caspase-7, and we map two putative caspase-7 cleavage sites to Asp residues at positions 266 and 344 of the ataxin-7 protein. Site-directed mutagenesis of these two caspase-7 cleavage sites in the polyQ-expanded form of ataxin-7 produces an ataxin-7 D266N/D344N protein that is resistant to caspase cleavage. Although ataxin-7 displays toxicity, forms nuclear aggregates, and represses transcription in human embryonic kidney 293T cells in a polyQ length-dependent manner, expression of the non-cleavable D266N/D344N form of polyQ-expanded ataxin-7 attenuated cell death, aggregate formation, and transcriptional interference. Expression of the caspase-7 truncation product of ataxin-7-69Q or -92Q, which removes the putative nuclear export signal and nuclear localization signals of ataxin-7, showed increased cellular toxicity. We also detected N-terminal polyQ-expanded ataxin-7 cleavage products in SCA7 transgenic mice similar in size to those generated by caspase-7 cleavage. In a SCA7 transgenic mouse model, recruitment of caspase-7 into the nucleus by polyQ-expanded ataxin-7 correlated with its activation. Our results, thus, suggest that proteolytic processing of ataxin-7 by caspase-7 may contribute to SCA7 disease pathogenesis.
Collapse
Affiliation(s)
- Jessica E Young
- Buck Institute for Age Research, Novato, California, 94945, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Latouche M, Lasbleiz C, Martin E, Monnier V, Debeir T, Mouatt-Prigent A, Muriel MP, Morel L, Ruberg M, Brice A, Stevanin G, Tricoire H. A conditional pan-neuronal Drosophila model of spinocerebellar ataxia 7 with a reversible adult phenotype suitable for identifying modifier genes. J Neurosci 2007; 27:2483-92. [PMID: 17344386 PMCID: PMC6672519 DOI: 10.1523/jneurosci.5453-06.2007] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Spinocerebellar ataxia 7 (SCA7) is a neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the ataxin 7 (ATXN7) protein, a member of a multiprotein complex involved in histone acetylation. We have created a conditional Drosophila model of SCA7 in which expression of truncated ATXN7 (ATXN7T) with a pathogenic polyQ expansion is induced in neurons in adult flies. In this model, mutant ATXN7T accumulated in neuronal intranuclear inclusions containing ubiquitin, the 19S proteasome subunit, and HSP70 (heat shock protein 70), as in patients. Aggregation was accompanied by a decrease in locomotion and lifespan but limited neuronal death. Disaggregation of the inclusions, when expression of expanded ATXN7T was stopped, correlated with improved locomotor function and increased lifespan, suggesting that the pathology may respond to treatment. Lifespan was then used as a quantitative marker in a candidate gene approach to validate the interest of the model and to identify generic modulators of polyQ toxicity and specific modifiers of SCA7. Several molecular pathways identified in this focused screen (proteasome function, unfolded protein stress, caspase-dependent apoptosis, and histone acetylation) were further studied in primary neuronal cultures. Sodium butyrate, a histone deacetylase inhibitor, improved the survival time of the neurons. This model is therefore a powerful tool for studying SCA7 and for the development of potential therapies for polyQ diseases.
Collapse
Affiliation(s)
- Morwena Latouche
- Institut National de la Santé et de la Recherche Médicale, Unité 679, Paris F-75013, France
- Université Pierre and Marie Curie–Paris 6, Institut Fédératif de Recherche de Neurosciences (IFR70), Unité Mixte de Recherche (UMR) S679, Group Hospitalier Pitié-Salpêtriére, Paris F-75013, France
| | - Christelle Lasbleiz
- Centre National de la Rechereche Scientifique, UMR 7592, Insititut Jacques Monod, Campus Universitaire de Jussieu, Paris F-75251, France, and
| | - Elodie Martin
- Institut National de la Santé et de la Recherche Médicale, Unité 679, Paris F-75013, France
- Université Pierre and Marie Curie–Paris 6, Institut Fédératif de Recherche de Neurosciences (IFR70), Unité Mixte de Recherche (UMR) S679, Group Hospitalier Pitié-Salpêtriére, Paris F-75013, France
| | - Véronique Monnier
- Centre National de la Rechereche Scientifique, UMR 7592, Insititut Jacques Monod, Campus Universitaire de Jussieu, Paris F-75251, France, and
| | - Thomas Debeir
- Institut National de la Santé et de la Recherche Médicale, Unité 679, Paris F-75013, France
- Université Pierre and Marie Curie–Paris 6, Institut Fédératif de Recherche de Neurosciences (IFR70), Unité Mixte de Recherche (UMR) S679, Group Hospitalier Pitié-Salpêtriére, Paris F-75013, France
| | - Annick Mouatt-Prigent
- Institut National de la Santé et de la Recherche Médicale, Unité 679, Paris F-75013, France
- Université Pierre and Marie Curie–Paris 6, Institut Fédératif de Recherche de Neurosciences (IFR70), Unité Mixte de Recherche (UMR) S679, Group Hospitalier Pitié-Salpêtriére, Paris F-75013, France
| | - Marie-Paule Muriel
- Institut National de la Santé et de la Recherche Médicale, Unité 679, Paris F-75013, France
- Université Pierre and Marie Curie–Paris 6, Institut Fédératif de Recherche de Neurosciences (IFR70), Unité Mixte de Recherche (UMR) S679, Group Hospitalier Pitié-Salpêtriére, Paris F-75013, France
| | - Lydie Morel
- Institut National de la Santé et de la Recherche Médicale, Unité 679, Paris F-75013, France
| | - Merle Ruberg
- Institut National de la Santé et de la Recherche Médicale, Unité 679, Paris F-75013, France
- Université Pierre and Marie Curie–Paris 6, Institut Fédératif de Recherche de Neurosciences (IFR70), Unité Mixte de Recherche (UMR) S679, Group Hospitalier Pitié-Salpêtriére, Paris F-75013, France
| | - Alexis Brice
- Institut National de la Santé et de la Recherche Médicale, Unité 679, Paris F-75013, France
- Université Pierre and Marie Curie–Paris 6, Institut Fédératif de Recherche de Neurosciences (IFR70), Unité Mixte de Recherche (UMR) S679, Group Hospitalier Pitié-Salpêtriére, Paris F-75013, France
- Assistance Publique–Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Départment de Génétique, Cytogénétique, et Embryologie, Paris F-75013, France
| | - Giovanni Stevanin
- Institut National de la Santé et de la Recherche Médicale, Unité 679, Paris F-75013, France
- Université Pierre and Marie Curie–Paris 6, Institut Fédératif de Recherche de Neurosciences (IFR70), Unité Mixte de Recherche (UMR) S679, Group Hospitalier Pitié-Salpêtriére, Paris F-75013, France
- Assistance Publique–Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Départment de Génétique, Cytogénétique, et Embryologie, Paris F-75013, France
| | - Hérvé Tricoire
- Centre National de la Rechereche Scientifique, UMR 7592, Insititut Jacques Monod, Campus Universitaire de Jussieu, Paris F-75251, France, and
| |
Collapse
|
48
|
Wang HL, He CY, Chou AH, Yeh TH, Chen YL, Li AH. Polyglutamine-expanded ataxin-7 decreases nuclear translocation of NF-κB p65 and impairs NF-κB activity by inhibiting proteasome activity of cerebellar neurons. Cell Signal 2007; 19:573-81. [PMID: 17005371 DOI: 10.1016/j.cellsig.2006.08.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Accepted: 08/13/2006] [Indexed: 01/10/2023]
Abstract
Our recent study indicated that polyglutamine-expanded ataxin-7-Q75 induced apoptotic death of cultured cerebellar neurons by downregulating Bcl-x(L) expression and activating mitochondrial apoptotic cascade. Mutant polyglutamine-expanded proteins are believed to impair the proteolytic function of ubiquitin-proteasome system by sequestering components of proteasomes. Proteasome degradation of IkappaBalpha permits nuclear translocation of NF-kappaB and is required for continuous NF-kappaB activity, which supports the survival of cultured cerebellar neurons by inducing Bcl-x(L) expression. Thus, we tested the hypothesis that mutant ataxin-7-Q75 causes proteasome dysfunction and impairs NF-kappaB activity, leading to reduced Bcl-x(L) expression, caspase activation and cerebellar neuronal death. EMSA assays indicate that DNA-binding activity of NF-kappaB was significantly decreased in cerebellar neurons expressing ataxin-7-Q75. Similar to mutant ataxin-7-Q75, NF-kappaB inhibitor APEQ induced cerebellar neuronal death by decreasing Bcl-x(L) expression and activating caspase-9. Mutant ataxin-7-Q75 inhibited the proteolytic activity of proteasomes in cerebellar neurons. Proteasome inhibitor MG132 also caused cerebellar neuronal death by decreasing Bcl-x(L) expression and activating caspase-9. Both ataxin-7-Q75 and MG132 caused the cytosolic accumulation of IkappaBalpha in cerebellar neurons. Mutant ataxin-7-Q75 or MG132 increased the cytosolic level of NF-kappaB p65 and decreased the nuclear NF-kappaB p65 level. Our study provides the evidence that polyglutamine-expanded ataxin-7-Q75 decreases nuclear translocation of NF-kappaB p65 and impairs NF-kappaB activity by inhibiting proteasome activity of cerebellar neurons.
Collapse
Affiliation(s)
- Hung-Li Wang
- Department of Physiology, Chang Gung University School of Medicine, Taiwan, ROC.
| | | | | | | | | | | |
Collapse
|
49
|
Woulfe JM. Abnormalities of the nucleus and nuclear inclusions in neurodegenerative disease: a work in progress. Neuropathol Appl Neurobiol 2007; 33:2-42. [PMID: 17239006 DOI: 10.1111/j.1365-2990.2006.00819.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are characterized pathologically by the abnormal accumulation of pathogenic protein species within the cell. Several neurodegenerative diseases feature intranuclear protein aggregation in the form of intranuclear inclusion bodies. Studies of these intranuclear inclusions are providing important clues regarding the cellular pathophysiology of these diseases, as exemplified by recent progress in defining the genetic basis of a subset of frontotemporal dementia cases. The precise role of intranuclear inclusion bodies in disease pathogenesis is currently a focus of debate. The present review provides an overview of the diverse family of neurodegenerative diseases in which nuclear inclusions form part of the neuropathological spectrum. In addition, current pathogenetic concepts relevant to these diseases will be reviewed and arguments for and against a protective role for intranuclear inclusions will be presented. The relationship of pathological intranuclear inclusions to functional intranuclear bodies will also be discussed. Finally, by analogy with pathological intranuclear inclusions, I will speculate on the possibility that intranuclear protein aggregation may represent a constitutive cellular protective mechanism occurring in neurons under physiological conditions.
Collapse
Affiliation(s)
- J M Woulfe
- Department of Pathology, The Ottawa Hospital, University of Ottawa, and Cancer Research Program, The Ottawa Health Research Institute, Ottawa, Canada.
| |
Collapse
|
50
|
Janer A, Martin E, Muriel MP, Latouche M, Fujigasaki H, Ruberg M, Brice A, Trottier Y, Sittler A. PML clastosomes prevent nuclear accumulation of mutant ataxin-7 and other polyglutamine proteins. ACTA ACUST UNITED AC 2006; 174:65-76. [PMID: 16818720 PMCID: PMC2064165 DOI: 10.1083/jcb.200511045] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The pathogenesis of spinocerebellar ataxia type 7 and other neurodegenerative polyglutamine (polyQ) disorders correlates with the aberrant accumulation of toxic polyQ-expanded proteins in the nucleus. Promyelocytic leukemia protein (PML) nuclear bodies are often present in polyQ aggregates, but their relation to pathogenesis is unclear. We show that expression of PML isoform IV leads to the formation of distinct nuclear bodies enriched in components of the ubiquitin-proteasome system. These bodies recruit soluble mutant ataxin-7 and promote its degradation by proteasome-dependent proteolysis, thus preventing the aggregate formation. Inversely, disruption of the endogenous nuclear bodies with cadmium increases the nuclear accumulation and aggregation of mutant ataxin-7, demonstrating their role in ataxin-7 turnover. Interestingly, β-interferon treatment, which induces the expression of endogenous PML IV, prevents the accumulation of transiently expressed mutant ataxin-7 without affecting the level of the endogenous wild-type protein. Therefore, clastosomes represent a potential therapeutic target for preventing polyQ disorders.
Collapse
Affiliation(s)
- Alexandre Janer
- Institut National de la Santé et de la Recherche Médicale U679, Neurologie et Thérapeutique Expérimentale, 75651 Paris Cedex 13, France.
| | | | | | | | | | | | | | | | | |
Collapse
|