1
|
Bellitto D, Bozzo M, Ravera S, Bertola N, Rosamilia F, Milia J, Barboro P, Vargas GC, Di Lisa D, Pastorino L, Lantieri F, Castagnola P, Iervasi E, Ponassi M, Profumo A, Tkachenko K, Rosano C, Candiani S, Bachetti T. A multi-omics approach reveals impaired lipid metabolism and oxidative stress in a zebrafish model of Alexander disease. Redox Biol 2025; 81:103544. [PMID: 40023981 PMCID: PMC11915002 DOI: 10.1016/j.redox.2025.103544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 03/04/2025] Open
Abstract
Alexander disease (AxD) is a rare leukodystrophy caused by heterozygous mutations in the GFAP gene. To date, several in vitro and in vivo models have been generated in an attempt to unravel the main mechanisms underlying this complex disease. However, none of these models is suitable for investigating the global dysregulation caused by AxD. To address this shortcoming, we have generated a stable transgenic zebrafish line (zAxD) carrying the human GFAP p.R239C mutation, which is associated with severe phenotypes of AxD type I patients. We then performed transcriptomics and proteomics analyses on the whole larvae of our zAxD model, confirming the involvement of several pathways such as the immune system response and inflammation, oxidative stress, extracellular matrix, lipoxidation and lipid metabolism, which were previously reported in more limited omic studies. Interestingly, new pathways emerged as well, including tyrosine and butanoate metabolic processes. Biochemical assays confirmed alterations in cell respiration and lipid metabolism as well as elevated oxidative stress. These findings confirm the reliability of the zAxD model to apply a whole-organism approach to investigate the molecular basis of the disease.
Collapse
Affiliation(s)
- Deianira Bellitto
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, Genova, Italy
| | - Matteo Bozzo
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, Genova, Italy
| | - Silvia Ravera
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Nadia Bertola
- IRCCS Ospedale Policlinico San Martino, Genova, Unità Patologia Clinica, Italy
| | - Francesca Rosamilia
- Bioinformatica Clinica, Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Jessica Milia
- Centro di Ricerca, Sviluppo e Studi Superiori in Sardegna (CRS4), Pula, Italy
| | - Paola Barboro
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Donatella Di Lisa
- Dipartimento di Informatica, Bioingegneria, Robotica e Ingegneria dei Sistemi, Università di Genova, Genova, Italy
| | - Laura Pastorino
- Dipartimento di Informatica, Bioingegneria, Robotica e Ingegneria dei Sistemi, Università di Genova, Genova, Italy
| | - Francesca Lantieri
- Dipartimento di Scienze della Salute, Università di Genova, Genova, Italy
| | - Patrizio Castagnola
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, Genova, Italy
| | - Erika Iervasi
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marco Ponassi
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Aldo Profumo
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | | | - Simona Candiani
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita, Università di Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | | |
Collapse
|
2
|
Lin NH, Jian WS, Perng MD. Deletions in Glial Fibrillary Acidic Protein Leading to Alterations in Intermediate Filament Assembly and Network Formation. Int J Mol Sci 2025; 26:1913. [PMID: 40076540 PMCID: PMC11900225 DOI: 10.3390/ijms26051913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Glial fibrillary acidic protein (GFAP) is classified as a type III intermediate filament protein predominantly expressed in mature astrocytes. It has the ability to self-assemble into 10 nm filaments in vitro, making it particularly valuable for elucidating the sequences essential for filament assembly. In this study, we created a series of deletion mutants targeting sequences in the N-terminal, C-terminal, and central rod domains to explore the sequences critical for the assembly of GFAP into 10 nm filaments. The impact of these deletions on filament formation was evaluated through in vitro assembly studies and transduction assays conducted with primary astrocytes. Our data revealed that deletions at the carboxy end resulted in abnormalities in either filament diameter calibration or lateral association, whereas deletions at the amino-terminal end significantly disrupted the filament assembly process, particularly restricting filament elongation. Furthermore, we discovered that the filament-forming sequences within the rod domain varied in their contributions to filament assembly and network formation. These findings enhance our understanding of the GFAP assembly process in vitro and provide a detailed mapping of the essential regions required for GFAP assembly. These insights hold significant implications for Alexander disease arising from deletion mutations in GFAP.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu 30043, Taiwan; (N.-H.L.); (W.-S.J.)
| | - Wan-Syuan Jian
- Institute of Molecular Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu 30043, Taiwan; (N.-H.L.); (W.-S.J.)
| | - Ming-Der Perng
- Institute of Molecular Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu 30043, Taiwan; (N.-H.L.); (W.-S.J.)
- School of Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu 30043, Taiwan
| |
Collapse
|
3
|
Bamford AR, Parkin GM, Corey-Bloom J, Thomas EA. Comparisons of neurodegenerative disease biomarkers across different biological fluids from patients with Huntington's disease. J Neurol 2025; 272:158. [PMID: 39849121 PMCID: PMC11759467 DOI: 10.1007/s00415-024-12785-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 01/25/2025]
Abstract
Fluid biomarkers play important roles in many aspects of neurodegenerative diseases, such as Huntington's disease (HD). However, a main question relates to how well levels of biomarkers measured in CSF are correlated with those measured in peripheral fluids, such as blood or saliva. In this study, we quantified levels of four neurodegenerative disease-related proteins, neurofilament light (NfL), total tau (t-tau), glial fibrillary acidic protein (GFAP) and YKL-40 in matched CSF, plasma and saliva samples from Huntingtin (HTT) gene-positive individuals (n = 21) using electrochemiluminescence assays. In addition, salivary levels of NfL, t-tau, and GFAP were quantified from a larger cohort (n = 95). We found both positive and negative correlations in the levels of these biomarkers among different biofluids. Most notably, in contrast to the significant positive correlations observed between CSF and plasma levels for NfL and GFAP, we detected significant negative correlations between the CSF and saliva levels of NfL and GFAP. With regard to clinical measures, both plasma and CSF levels of NfL were significantly positively correlated with Total Motor Score and chorea, whereas saliva levels of NfL showed significant correlations in the opposite direction. Additional correlations between salivary biomarkers with clinical data, adjusting for age, sex and CAG repeat length, confirmed that salivary NfL was significantly negatively associated with chorea scores in manifest HD, but not premanifest (PM), individuals. In contrast, salivary t-tau was positively associated with measures of cognition in PM participants. These findings suggest that salivary levels of NfL and t-tau proteins may exemplify non-invasive biomarkers for disease symptoms at different stages of illness. Further, these findings highlight the notion that different forms of disease proteins exist in different biological fluids.
Collapse
Affiliation(s)
- Alison R Bamford
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
- Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA
| | - Georgia M Parkin
- Phoenix Australia - Centre for Posttraumatic Mental Health, Department of Psychiatry, University of Melbourne, Parkville, VIC, Australia
| | - Jody Corey-Bloom
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Elizabeth A Thomas
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA.
- Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA.
- Department of Neurosciences, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
4
|
Zhang J, Zhang J, Yang C. Autophagy in brain tumors: molecular mechanisms, challenges, and therapeutic opportunities. J Transl Med 2025; 23:52. [PMID: 39806481 PMCID: PMC11727735 DOI: 10.1186/s12967-024-06063-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
Autophagy is responsible for maintaining cellular balance and ensuring survival. Autophagy plays a crucial role in the development of diseases, particularly human cancers, with actions that can either promote survival or induce cell death. However, brain tumors contribute to high levels of both mortality and morbidity globally, with resistance to treatments being acquired due to genetic mutations and dysregulation of molecular mechanisms, among other factors. Hence, having knowledge of the role of molecular processes in the advancement of brain tumors is enlightening, and the current review specifically examines the role of autophagy. The discussion would focus on the molecular pathways that control autophagy in brain tumors, and its dual role as a tumor suppressor and a supporter of tumor survival. Autophagy can control the advancement of different types of brain tumors like glioblastoma, glioma, and ependymoma, demonstrating its potential for treatment. Autophagy mechanisms can influence metastasis and drug resistance in glioblastoma, and there is a complex interplay between autophagy and cellular responses to stress like hypoxia and starvation. Autophagy can inhibit the growth of brain tumors by promoting apoptosis. Hence, focusing on autophagy could offer fresh perspectives on creating successful treatments.
Collapse
Affiliation(s)
- Jiarui Zhang
- Department of Pathology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinan Zhang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| | - Chen Yang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| |
Collapse
|
5
|
Matusova Z, Dykstra W, de Pablo Y, Zetterdahl OG, Canals I, van Gelder CAGH, Vos HR, Pérez-Sala D, Kubista M, Abaffy P, Ahlenius H, Valihrach L, Hol EM, Pekny M. Aberrant neurodevelopment in human iPS cell-derived models of Alexander disease. Glia 2025; 73:57-79. [PMID: 39308436 DOI: 10.1002/glia.24618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 12/21/2024]
Abstract
Alexander disease (AxD) is a rare and severe neurodegenerative disorder caused by mutations in glial fibrillary acidic protein (GFAP). While the exact disease mechanism remains unknown, previous studies suggest that mutant GFAP influences many cellular processes, including cytoskeleton stability, mechanosensing, metabolism, and proteasome function. While most studies have primarily focused on GFAP-expressing astrocytes, GFAP is also expressed by radial glia and neural progenitor cells, prompting questions about the impact of GFAP mutations on central nervous system (CNS) development. In this study, we observed impaired differentiation of astrocytes and neurons in co-cultures of astrocytes and neurons, as well as in neural organoids, both generated from AxD patient-derived induced pluripotent stem (iPS) cells with a GFAPR239C mutation. Leveraging single-cell RNA sequencing (scRNA-seq), we identified distinct cell populations and transcriptomic differences between the mutant GFAP cultures and a corrected isogenic control. These findings were supported by results obtained with immunocytochemistry and proteomics. In co-cultures, the GFAPR239C mutation resulted in an increased abundance of immature cells, while in unguided neural organoids and cortical organoids, we observed altered lineage commitment and reduced abundance of astrocytes. Gene expression analysis revealed increased stress susceptibility, cytoskeletal abnormalities, and altered extracellular matrix and cell-cell communication patterns in the AxD cultures, which also exhibited higher cell death after stress. Overall, our results point to altered cell differentiation in AxD patient-derived iPS-cell models, opening new avenues for AxD research.
Collapse
Affiliation(s)
- Zuzana Matusova
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Werner Dykstra
- Department of Translational Neuroscience, University Medical Centre Utrecht Brain Centre, Utrecht University, Utrecht, The Netherlands
| | - Yolanda de Pablo
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Oskar G Zetterdahl
- Stem Cells, Aging and Neurodegeneration Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
- Glial and Neuronal Biology Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Isaac Canals
- Glial and Neuronal Biology Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Metabolism, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
- ITINERARE-Innovative therapies in rare diseases, University Research Priority Program, University of Zurich, Zurich, Switzerland
| | - Charlotte A G H van Gelder
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Harmjan R Vos
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Henrik Ahlenius
- Stem Cells, Aging and Neurodegeneration Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Centre Utrecht Brain Centre, Utrecht University, Utrecht, The Netherlands
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
6
|
Hol EM, Dykstra W, Chevalier J, Cuadrado E, Bugiani M, Aronica E, Verkhratsky A. Neuroglia in leukodystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:159-175. [PMID: 40148043 DOI: 10.1016/b978-0-443-19102-2.00032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Leukodystrophies are a heterogeneous group of rare genetic neurologic disorders characterized by white matter degeneration resulting from mutations affecting glial cells. This review focuses on the primary subtypes-astroglial, oligodendroglial, and microglial leukodystrophies-offering a detailed description of their neuropathologic features and clinical manifestations. It delves into key aspects of the pathogenesis, emphasizing the distinct cellular mechanisms that drive white matter damage. Advances in disease modeling, including the development of animal models with pathologic gene expressions and patient-derived iPS-cell models, have significantly enhanced our understanding of these rare disorders. Insights into the roles of different glial cell types highlight the complexity of leukodystrophies and provide a foundation for the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Werner Dykstra
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Juliette Chevalier
- Department of Child Neurology and Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Eloy Cuadrado
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marianna Bugiani
- Department of Child Neurology and Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
7
|
Grossi A, Rosamilia F, Carestiato S, Salsano E, Ceccherini I, Bachetti T. A systematic review and meta-analysis of GFAP gene variants in Alexander disease. Sci Rep 2024; 14:24341. [PMID: 39420046 PMCID: PMC11487261 DOI: 10.1038/s41598-024-75383-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
Alexander disease (ALXDRD) is a rare neurodegenerative disorder of astrocytes resulting from pathogenic variants in the GFAP gene. The genotype-phenotype correlation remains elusive due to the variable expressivity of clinical manifestations. In an attempt to clarify the effects of GFAP variants in ALXDRD, numerous studies were collected and analyzed. In particular, we systematically searched for GFAP variants associated with ALXDRD and collected information on the location within the gene and protein, prediction of deleteriousness/pathogenicity, occurrence, sex and country of origin of patients, DNA source, genetic testing, and clinical signs. To identify possible associations, statistical analyses and meta-analyses were applied, thus revealing a higher than expected percentage of adult patients with ALXDRD. Furthermore, substitution of Arginine, the most frequently altered residue among the 550 predominantly missense causative GFAP variants collected, were mostly de novo and more prevalent in early-onset forms of ALXDRD. The effect of defective splicing in modifying the impact of GFAP variants on the age of onset of ALXDRD was also postulated after evaluating the distribution of the corresponding deleterious predictive values. In conclusion, not only previously unrecognized genotype-phenotype correlations were revealed in ALXDRD, but also subtle mechanisms could explain the variable manifestations of the ALXDRD clinical phenotype.
Collapse
Affiliation(s)
- Alice Grossi
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, Genoa, 16147, Italy
| | - Francesca Rosamilia
- Clinical Bioinformatics, IRCCS Istituto Giannina Gaslini, Genoa, 16147, Italy
| | - Silvia Carestiato
- Department of Neurosciences, Rita Levi Montalcini University of Turin, Turin, 10126, Italy
| | - Ettore Salsano
- SC Malattie Neurologiche Rare, Fondazione IRCCS Istituto Neurologico C. Besta, Milano, Italy
| | - Isabella Ceccherini
- Laboratory of Genetics and Genomics of Rare Diseases, IRCCS Istituto Giannina Gaslini, Genoa, 16147, Italy.
- UOSD Laboratory of Genetics and Genomics of rare Diseases, IRCCS Istituto Giannina gaslini, Via G Gaslini, 5, Genova, 16148, Italy.
| | | |
Collapse
|
8
|
Bailly C. Covalent binding of withanolides to cysteines of protein targets. Biochem Pharmacol 2024; 226:116405. [PMID: 38969301 DOI: 10.1016/j.bcp.2024.116405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/26/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Withanolides represent an important category of natural products with a steroidal lactone core. Many of them contain an α,β-unsaturated carbonyl moiety with a high reactivity toward sulfhydryl groups, including protein cysteine thiols. Different withanolides endowed with marked antitumor and anti-inflammatory have been shown to form stable covalent complexes with exposed cysteines present in the active site of oncogenic kinases (BTK, IKKβ, Zap70), metabolism enzymes (Prdx-1/6, Pin1, PHGDH), transcription factors (Nrf2, NFκB, C/EBPβ) and other structural and signaling molecules (GFAP, β-tubulin, p97, Hsp90, vimentin, Mpro, IPO5, NEMO, …). The present review analyzed the covalent complexes formed through Michael addition alkylation reactions between six major withanolides (withaferin A, physalin A, withangulatin A, 4β-hydroxywithanolide E, withanone and tubocapsanolide A) and key cysteine residues of about 20 proteins and the resulting biological effects. The covalent conjugation of the α,β-unsaturated carbonyl system of withanolides with reactive protein thiols can occur with a large set of soluble and membrane proteins. It points to a general mechanism, well described with the leading natural product withaferin A, but likely valid for most withanolides harboring a reactive (electrophilic) enone moiety susceptible to react covalently with cysteinyl residues of proteins. The multiplicity of reactive proteins should be taken into account when studying the mechanism of action of new withanolides. Proteomic and network analyses shall be implemented to capture and compare the cysteine covalent-binding map for the major withanolides, so as to identify the protein targets at the origin of their activity and/or unwanted effects. Screening of the cysteinome will help understanding the mechanism of action and designing cysteine-reactive electrophilic drug candidates.
Collapse
Affiliation(s)
- Christian Bailly
- CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, OncoLille Institute, University of Lille, F-59000 Lille, France; Institute of Pharmaceutical Chemistry Albert Lespagnol (ICPAL), Faculty of Pharmacy, University of Lille, F-59006 Lille, France; OncoWitan, Scientific Consulting Office, F-59290 Lille, France.
| |
Collapse
|
9
|
Lin NH, Jian WS, Snider N, Perng MD. Glial fibrillary acidic protein is pathologically modified in Alexander disease. J Biol Chem 2024; 300:107402. [PMID: 38782207 PMCID: PMC11259701 DOI: 10.1016/j.jbc.2024.107402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Here, we describe pathological events potentially involved in the disease pathogenesis of Alexander disease (AxD). This is a primary genetic disorder of astrocyte caused by dominant gain-of-function mutations in the gene coding for an intermediate filament protein glial fibrillary acidic protein (GFAP). Pathologically, this disease is characterized by the upregulation of GFAP and its accumulation as Rosenthal fibers. Although the genetic basis linking GFAP mutations with Alexander disease has been firmly established, the initiating events that promote GFAP accumulation and the role of Rosenthal fibers (RFs) in the disease process remain unknown. Here, we investigate the hypothesis that disease-associated mutations promote GFAP aggregation through aberrant posttranslational modifications. We found high molecular weight GFAP species in the RFs of AxD brains, indicating abnormal GFAP crosslinking as a prominent pathological feature of this disease. In vitro and cell-based studies demonstrate that cystine-generating mutations promote GFAP crosslinking by cysteine-dependent oxidation, resulting in defective GFAP assembly and decreased filament solubility. Moreover, we found GFAP was ubiquitinated in RFs of AxD patients and rodent models, supporting this modification as a critical factor linked to GFAP aggregation. Finally, we found that arginine could increase the solubility of aggregation-prone mutant GFAP by decreasing its ubiquitination and aggregation. Our study suggests a series of pathogenic events leading to AxD, involving interplay between GFAP aggregation and abnormal modifications by GFAP ubiquitination and oxidation. More important, our findings provide a basis for investigating new strategies to treat AxD by targeting abnormal GFAP modifications.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Wan-Syuan Jian
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Natasha Snider
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ming-Der Perng
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan; School of Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
10
|
Pajares MA, Hernández-Gerez E, Pekny M, Pérez-Sala D. Alexander disease: the road ahead. Neural Regen Res 2023; 18:2156-2160. [PMID: 37056123 DOI: 10.4103/1673-5374.369097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023] Open
Abstract
Alexander disease is a rare neurodegenerative disorder caused by mutations in the glial fibrillary acidic protein, a type III intermediate filament protein expressed in astrocytes. Both early (infantile or juvenile) and adult onsets of the disease are known and, in both cases, astrocytes present characteristic aggregates, named Rosenthal fibers. Mutations are spread along the glial fibrillary acidic protein sequence disrupting the typical filament network in a dominant manner. Although the presence of aggregates suggests a proteostasis problem of the mutant forms, this behavior is also observed when the expression of wild-type glial fibrillary acidic protein is increased. Additionally, several isoforms of glial fibrillary acidic protein have been described to date, while the impact of the mutations on their expression and proportion has not been exhaustively studied. Moreover, the posttranslational modification patterns and/or the protein-protein interaction networks of the glial fibrillary acidic protein mutants may be altered, leading to functional changes that may modify the morphology, positioning, and/or the function of several organelles, in turn, impairing astrocyte normal function and subsequently affecting neurons. In particular, mitochondrial function, redox balance and susceptibility to oxidative stress may contribute to the derangement of glial fibrillary acidic protein mutant-expressing astrocytes. To study the disease and to develop putative therapeutic strategies, several experimental models have been developed, a collection that is in constant growth. The fact that most cases of Alexander disease can be related to glial fibrillary acidic protein mutations, together with the availability of new and more relevant experimental models, holds promise for the design and assay of novel therapeutic strategies.
Collapse
Affiliation(s)
- María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| | - Elena Hernández-Gerez
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; University of Newcastle, Newcastle, NSW, and the Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| |
Collapse
|
11
|
Goldman JE. Alzheimer Type I Astrocytes: Still Mysterious Cells. J Neuropathol Exp Neurol 2022; 81:588-595. [PMID: 35689655 DOI: 10.1093/jnen/nlac043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Over 100 years ago, von Hösslein and Alzheimer described enlarged and multinucleated astrocytes in the brains of patients with Wilson disease. These odd astrocytes, now well known to neuropathologists, are present in a large variety of neurological disorders, and yet the mechanisms underlying their generation and their functional attributes are still not well understood. They undergo abnormal mitoses and fail to accomplish cytokinesis, resulting in multinucleation. Oxidative stress, hypoxia, and inflammation may be contributing pathologies to generate these astrocytes. The abnormal mitoses occur from changes in cell shape, the accumulation of cytoplasmic proteins, and the mislocalization of many of the important molecules whose coordination is necessary for proper mitotic spindle formation. Modern technologies will be able to characterize their abnormalities and solve century old questions of their form and function.
Collapse
Affiliation(s)
- James E Goldman
- From the Division of Neuropathology, Department of Pathology & Cell Biology, Columbia University Vagelos College of Physicians and Surgeons and The Taub Institute for Research on Alzheimer's Disease and Aging, NY-Presbyterian Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
12
|
Yang AW, Lin NH, Yeh TH, Snider N, Perng MD. Effects of Alexander disease-associated mutations on the assembly and organization of GFAP intermediate filaments. Mol Biol Cell 2022; 33:ar69. [PMID: 35511821 PMCID: PMC9635275 DOI: 10.1091/mbc.e22-01-0013] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/30/2022] [Accepted: 04/25/2022] [Indexed: 02/02/2023] Open
Abstract
Alexander disease is a primary genetic disorder of astrocytes caused by dominant mutations in the gene encoding glial fibrillary acidic protein (GFAP). How single-amino-acid changes can lead to cytoskeletal catastrophe and brain degeneration remains poorly understood. In this study, we have analyzed 14 missense mutations located in the GFAP rod domain to investigate how these mutations affect in vitro filament assembly. Whereas the internal rod mutants assembled into filaments that were shorter than those of wild type, the rod end mutants formed structures with one or more of several atypical characteristics, including short filament length, irregular width, roughness of filament surface, and filament aggregation. When transduced into primary astrocytes, GFAP mutants with in vitro assembly defects usually formed cytoplasmic aggregates, which were more resistant to biochemical extraction. The resistance of GFAP to solubilization was also observed in brain tissues of patients with Alexander disease, in which a significant proportion of insoluble GFAP were accumulated in Rosenthal fiber fractions. These findings provide clinically relevant evidence that link GFAP assembly defects to disease pathology at the tissue level and suggest that altered filament assembly and properties as a result of GFAP mutation are critical initiating factors for the pathogenesis of Alexander disease.
Collapse
Affiliation(s)
- Ai-Wen Yang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ni-Hsuan Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ting-Hung Yeh
- Department of Medical Science, College of Life Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Natasha Snider
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Ming-Der Perng
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Medical Science, College of Life Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
13
|
Becerra-Hernández LV, Escobar-Betancourt MI, Pimienta-Jiménez HJ, Buriticá E. Crystallin Alpha-B Overexpression as a Possible Marker of Reactive Astrogliosis in Human Cerebral Contusions. Front Cell Neurosci 2022; 16:838551. [PMID: 35360493 PMCID: PMC8963874 DOI: 10.3389/fncel.2022.838551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The pathophysiology of traumatic brain injury (TBI) has not yet been fully elucidated. Crystallin alpha-B (CRYAB) is a molecular chaperone that apparently tries to stabilize the rapid thickening of the intermediate filaments of glial fibrillary acidic protein (GFAP) during the process of reactive astrogliosis in response to TBI. Previous analyses of the gene expression profile in human brain contusion tissue showed us an exacerbated CRYAB overexpression. Here, we used 3, 3’-diaminobenzidine (DAB) immunohistochemistry and immunofluorescence to verify CRYAB overexpression and to describe its expression and distribution in samples of contused cortical tissue derived from emergency decompressive surgery after severe TBI. The histological expression of CRYAB was mainly seen in subcortical white matter astrocytes of injured tissue. Most of the cells that overexpressed GFAP in the analyzed tissue also overexpressed CRYAB, a finding corroborated by the co-localization of the two markers. The only difference was the presence of a few pyramidal neurons that expressed CRYAB in layer V of the cerebral cortex. The selective vulnerability of layer V of the cerebral cortex during TBI could explain the expression of CRYAB in neurons of this cortical layer. Our results indicate a parallel behavior in the cellular expression of CRYAB and GFAP during the subacute response to TBI. These results lead us to postulate CRYAB as a possible marker of reactive astrogliosis in contused cortical tissue.
Collapse
|
14
|
Zarekiani P, Nogueira Pinto H, Hol EM, Bugiani M, de Vries HE. The neurovascular unit in leukodystrophies: towards solving the puzzle. Fluids Barriers CNS 2022; 19:18. [PMID: 35227276 PMCID: PMC8887016 DOI: 10.1186/s12987-022-00316-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
The neurovascular unit (NVU) is a highly organized multicellular system localized in the brain, formed by neuronal, glial (astrocytes, oligodendrocytes, and microglia) and vascular (endothelial cells and pericytes) cells. The blood-brain barrier, a complex and dynamic endothelial cell barrier in the brain microvasculature that separates the blood from the brain parenchyma, is a component of the NVU. In a variety of neurological disorders, including Alzheimer's disease, multiple sclerosis, and stroke, dysfunctions of the NVU occurs. There is, however, a lack of knowledge regarding the NVU function in leukodystrophies, which are rare monogenic disorders that primarily affect the white matter. Since leukodystrophies are rare diseases, human brain tissue availability is scarce and representative animal models that significantly recapitulate the disease are difficult to develop. The introduction of human induced pluripotent stem cells (hiPSC) now makes it possible to surpass these limitations while maintaining the ability to work in a biologically relevant human context and safeguarding the genetic background of the patient. This review aims to provide further insights into the NVU functioning in leukodystrophies, with a special focus on iPSC-derived models that can be used to dissect neurovascular pathophysiology in these diseases.
Collapse
Affiliation(s)
- Parand Zarekiani
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Henrique Nogueira Pinto
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Hagemann TL. Alexander disease: models, mechanisms, and medicine. Curr Opin Neurobiol 2022; 72:140-147. [PMID: 34826654 PMCID: PMC8901527 DOI: 10.1016/j.conb.2021.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 02/03/2023]
Abstract
Alexander disease is a primary disorder of astrocytes caused by gain-of-function mutations in the gene for glial fibrillary acidic protein (GFAP), which lead to protein aggregation and a reactive astrocyte response, with devastating effects on the central nervous system. Over the past two decades since the discovery of GFAP as the culprit, several cellular and animal models have been generated, and much has been learned about underlying mechanisms contributing to the disease. Despite these efforts, many aspects of Alexander disease have remained enigmatic, particularly the initiating events in GFAP accumulation and astrocyte pathology, the relation between astrocyte dysfunction and myelin deficits, and the variability in age of onset and disease severity. More recent work in both old and new models has begun to address these complex questions and identify new therapeutics that finally offer the promise of effective treatment.
Collapse
Affiliation(s)
- Tracy L. Hagemann
- Waisman Center, University of Wisconsin – Madison, 1500 Highland Ave, Madison, WI 53705
| |
Collapse
|
16
|
Abstract
Fifty years have passed since the discovery of glial fibrillary acidic protein (GFAP) by Lawrence Eng and colleagues. Now recognized as a member of the intermediate filament family of proteins, it has become a subject for study in fields as diverse as structural biology, cell biology, gene expression, basic neuroscience, clinical genetics and gene therapy. This review covers each of these areas, presenting an overview of current understanding and controversies regarding GFAP with the goal of stimulating continued study of this fascinating protein.
Collapse
Affiliation(s)
- Albee Messing
- Waisman Center, University of Wisconsin-Madison.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison
| | - Michael Brenner
- Department of Neurobiology, University of Alabama-Birmingham
| |
Collapse
|
17
|
Saito K, Shigetomi E, Koizumi S. [Alexander disease: diversity of cell population and interactions between neuron and glia]. Nihon Yakurigaku Zasshi 2021; 156:239-243. [PMID: 34193704 DOI: 10.1254/fpj.21028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Alexander disease (AxD) is a rare neurodegenerative disorder caused by the mutations in glial fibrillary acidic protein (GFAP) gene. Rosenthal fiber formations in astrocytes are the pathological hallmarks of AxD. Astrocyte dysfunction in the AxD brain is considered to be involved in its pathogenesis. We have previously reported that in AxD model mice aberrant Ca2+ signals in astrocytes were associated with the upregulation of reactive phenotype. Reactive astrocytes are conditions that lead to morphological, functional, and molecular changes by responding to various pathological insults (trauma, inflammation, ischemia), and environmental stimuli. Recent technological advances in single-cell gene expression analysis have revealed that astrocytes have heterogeneity by indicating that they form sub population with different characteristics depending on the brain region, the growth development, aging stage, and the pathological condition. AxD astrocytes are also thought to constitute a heterogeneous population with diverse properties and functions. Moreover, it is presumed that AxD pathogenesis occur due to interactions with neurons and other glial cells, as well as the microenvironment in tissues. Research strategies based on these perspectives will help us understand AxD pathology better and may lead to the elucidation of disease modifiers and clinical diversity.
Collapse
Affiliation(s)
- Kozo Saito
- Department of Neuropharmcology, Interdisciplinary Graduate School of Medicine
| | - Eiji Shigetomi
- Department of Neuropharmcology, Interdisciplinary Graduate School of Medicine
| | - Schuichi Koizumi
- Department of Neuropharmcology, Interdisciplinary Graduate School of Medicine
| |
Collapse
|
18
|
Song X, Jiang J, Tian W, Zhan F, Zhu Z, Li B, Tang H, Cao L. A report of two cases of bulbospinal form Alexander disease and preliminary exploration of the disease. Mol Med Rep 2021; 24:572. [PMID: 34109421 PMCID: PMC8201446 DOI: 10.3892/mmr.2021.12211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/10/2021] [Indexed: 11/05/2022] Open
Abstract
Alexander disease (AxD) is a cerebral white matter disease affecting a wide range of ages, from infants to adults. In the present study, two cases of bulbospinal form AxD were reported, and a preliminary exploration of AxD was conducted thorough clinical, functional magnetic resonance imaging (fMRI) and functional analyses. In total, two de novo mutations in the glial fibrillary acidic protein (GFAP) gene (c.214G>A and c.1235C>T) were identified in unrelated patients (one in each patient). Both patients showed increased regional neural activity and functional connectivity in the cerebellum and posterior parietal cortex according to fMRI analysis. Notably, grey matter atrophy was discovered in the patient with c.214G>A variant. Functional experiments revealed aberrant accumulation of mutant GFAP and decreased solubility of c.1235C>T variant. Under pathological conditions, autophagic flux was activated for GFAP aggregate degradation. Moreover, transcriptional data of AxD and healthy human brain samples were obtained from the Gene Expression Omnibus database. Gene set enrichment analysis revealed an upregulation of immune‑related responses and downregulation of ion transport, synaptic transmission and neurotransmitter homeostasis. Enrichment analysis of cell‑specific differentially expressed genes also indicated a marked inflammatory environment in AxD. Overall, the clinical features of the two patients with bulbospinal form AxD were thoroughly described. To the best of our knowledge, the brain atrophy pattern and spontaneous brain functional network activity of patients with AxD were explored for the first time. Cytological experiments provided evidence of the pathogenicity of the identified variants. Furthermore, bioinformatics analysis found that inflammatory immune‑related reactions may play a critical role in AxD, which may be conducive to the understanding of this disease.
Collapse
Affiliation(s)
- Xiaoxuan Song
- Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Jingwen Jiang
- Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Wotu Tian
- Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Feixia Zhan
- Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Zeyu Zhu
- Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Binyin Li
- Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Huidong Tang
- Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Li Cao
- Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
19
|
Zarekiani P, Breur M, Wolf NI, de Vries HE, van der Knaap MS, Bugiani M. Pathology of the neurovascular unit in leukodystrophies. Acta Neuropathol Commun 2021; 9:103. [PMID: 34082828 PMCID: PMC8173888 DOI: 10.1186/s40478-021-01206-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/24/2021] [Indexed: 01/20/2023] Open
Abstract
The blood-brain barrier is a dynamic endothelial cell barrier in the brain microvasculature that separates the blood from the brain parenchyma. Specialized brain endothelial cells, astrocytes, neurons, microglia and pericytes together compose the neurovascular unit and interact to maintain blood-brain barrier function. A disturbed brain barrier function is reported in most common neurological disorders and may play a role in disease pathogenesis. However, a comprehensive overview of how the neurovascular unit is affected in a wide range of rare disorders is lacking. Our aim was to provide further insights into the neuropathology of the neurovascular unit in leukodystrophies to unravel its potential pathogenic role in these diseases. Leukodystrophies are monogenic disorders of the white matter due to defects in any of its structural components. Single leukodystrophies are exceedingly rare, and availability of human tissue is unique. Expression of selective neurovascular unit markers such as claudin-5, zona occludens 1, laminin, PDGFRβ, aquaporin-4 and α-dystroglycan was investigated in eight different leukodystrophies using immunohistochemistry. We observed tight junction rearrangements, indicative of endothelial dysfunction, in five out of eight assessed leukodystrophies of different origin and an altered aquaporin-4 distribution in all. Aquaporin-4 redistribution indicates a general astrocytic dysfunction in leukodystrophies, even in those not directly related to astrocytic pathology or without prominent reactive astrogliosis. These findings provide further evidence for dysfunction in the orchestration of the neurovascular unit in leukodystrophies and contribute to a better understanding of the underlying disease mechanism.
Collapse
Affiliation(s)
- Parand Zarekiani
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, de Boelelaan 1117, 1081HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marjolein Breur
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Nicole I. Wolf
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Helga E. de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marjo S. van der Knaap
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, de Boelelaan 1117, 1081HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Uranga J, Hasecke L, Proppe J, Fingerhut J, Mata RA. Theoretical Studies of the Acid-Base Equilibria in a Model Active Site of the Human 20S Proteasome. J Chem Inf Model 2021; 61:1942-1953. [PMID: 33719420 DOI: 10.1021/acs.jcim.0c01459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The 20S proteasome is a macromolecule responsible for the chemical step in the ubiquitin-proteasome system of degrading unnecessary and unused proteins of the cell. It plays a central role both in the rapid growth of cancer cells and in viral infection cycles. Herein, we present a computational study of the acid-base equilibria in an active site of the human proteasome (caspase-like), an aspect which is often neglected despite the crucial role protons play in the catalysis. As example substrates, we take the inhibition by epoxy- and boronic acid-containing warheads. We have combined cluster quantum mechanical calculations, replica exchange molecular dynamics, and Bayesian optimization of nonbonded potential terms in the inhibitors. In relation to the latter, we propose an easily scalable approach for the reevaluation of nonbonded potentials making use of the hybrid quantum mechanics molecular mechanics dynamics information. Our results show that coupled acid-base equilibria need to be considered when modeling the inhibition mechanism. The coupling between a neighboring lysine and the reacting threonine is not affected by the presence of the studied inhibitors.
Collapse
Affiliation(s)
- Jon Uranga
- Institute of Physical Chemistry, University of Göttingen, Tammannstrasse 6, 37077 Göttingen, Germany
| | - Lukas Hasecke
- Institute of Physical Chemistry, University of Göttingen, Tammannstrasse 6, 37077 Göttingen, Germany
| | - Jonny Proppe
- Institute of Physical Chemistry, University of Göttingen, Tammannstrasse 6, 37077 Göttingen, Germany
| | - Jan Fingerhut
- Institute of Physical Chemistry, University of Göttingen, Tammannstrasse 6, 37077 Göttingen, Germany
| | - Ricardo A Mata
- Institute of Physical Chemistry, University of Göttingen, Tammannstrasse 6, 37077 Göttingen, Germany
| |
Collapse
|
21
|
D'Mello SR. MECP2 and the Biology of MECP2 Duplication Syndrome. J Neurochem 2021; 159:29-60. [PMID: 33638179 DOI: 10.1111/jnc.15331] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/21/2021] [Accepted: 02/18/2021] [Indexed: 11/27/2022]
Abstract
MECP2 duplication syndrome (MDS), a rare X-linked genomic disorder affecting predominantly males, is caused by duplication of the chromosomal region containing the methyl CpG binding protein-2 (MECP2) gene, which encodes methyl-CpG-binding protein 2 (MECP2), a multi-functional protein required for proper brain development and maintenance of brain function during adulthood. Disease symptoms include severe motor and cognitive impairment, delayed or absent speech development, autistic features, seizures, ataxia, recurrent respiratory infections and shortened lifespan. The cellular and molecular mechanisms by which a relatively modest increase in MECP2 protein causes such severe disease symptoms are poorly understood and consequently there are no treatments available for this fatal disorder. This review summarizes what is known to date about the structure and complex regulation of MECP2 and its many functions in the developing and adult brain. Additionally, recent experimental findings on the cellular and molecular underpinnings of MDS based on cell culture and mouse models of the disorder are reviewed. The emerging picture from these studies is that MDS is a neurodegenerative disorder in which neurons die in specific parts of the central nervous system, including the cortex, hippocampus, cerebellum and spinal cord. Neuronal death likely results from astrocytic dysfunction, including a breakdown of glutamate homeostatic mechanisms. The role of elevations in the expression of glial acidic fibrillary protein (GFAP) in astrocytes and the microtubule-associated protein, Tau, in neurons to the pathogenesis of MDS is discussed. Lastly, potential therapeutic strategies to potentially treat MDS are discussed.
Collapse
|
22
|
de Waard DM, Bugiani M. Astrocyte-Oligodendrocyte-Microglia Crosstalk in Astrocytopathies. Front Cell Neurosci 2020; 14:608073. [PMID: 33328899 PMCID: PMC7710860 DOI: 10.3389/fncel.2020.608073] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022] Open
Abstract
Defective astrocyte function due to a genetic mutation can have major consequences for microglia and oligodendrocyte physiology, which in turn affects the white matter integrity of the brain. This review addresses the current knowledge on shared and unique pathophysiological mechanisms of astrocytopathies, including vanishing white matter, Alexander disease, megalencephalic leukoencephalopathy with subcortical cysts, Aicardi-Goutières syndrome, and oculodentodigital dysplasia. The mechanisms of disease include protein accumulation, unbalanced secretion of extracellular matrix proteins, pro- and anti-inflammatory molecules, cytokines and chemokines by astrocytes, as well as an altered gap junctional network and a changed ionic and nutrient homeostasis. Interestingly, the extent to which astrogliosis and microgliosis are present in these astrocytopathies is highly variable. An improved understanding of astrocyte-microglia-oligodendrocyte crosstalk might ultimately lead to the identification of druggable targets for these, currently untreatable, severe conditions.
Collapse
Affiliation(s)
| | - Marianna Bugiani
- Department of Pathology, VU Medical center, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
23
|
Sjöqvist M, Antfolk D, Suarez-Rodriguez F, Sahlgren C. From structural resilience to cell specification - Intermediate filaments as regulators of cell fate. FASEB J 2020; 35:e21182. [PMID: 33205514 PMCID: PMC7839487 DOI: 10.1096/fj.202001627r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 12/18/2022]
Abstract
During the last decades intermediate filaments (IFs) have emerged as important regulators of cellular signaling events, ascribing IFs with functions beyond the structural support they provide. The organ and developmental stage‐specific expression of IFs regulate cell differentiation within developing or remodeling tissues. Lack of IFs causes perturbed stem cell differentiation in vasculature, intestine, nervous system, and mammary gland, in transgenic mouse models. The aberrant cell fate decisions are caused by deregulation of different stem cell signaling pathways, such as Notch, Wnt, YAP/TAZ, and TGFβ. Mutations in genes coding for IFs cause an array of different diseases, many related to stem cell dysfunction, but the molecular mechanisms remain unresolved. Here, we provide a comprehensive overview of how IFs interact with and regulate the activity, localization and function of different signaling proteins in stem cells, and how the assembly state and PTM profile of IFs may affect these processes. Identifying when, where and how IFs and cell signaling congregate, will expand our understanding of IF‐linked stem cell dysfunction during development and disease.
Collapse
Affiliation(s)
- Marika Sjöqvist
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Daniel Antfolk
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Freddy Suarez-Rodriguez
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Cecilia Sahlgren
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
24
|
Trujillo-Estrada L, Gomez-Arboledas A, Forner S, Martini AC, Gutierrez A, Baglietto-Vargas D, LaFerla FM. Astrocytes: From the Physiology to the Disease. Curr Alzheimer Res 2020; 16:675-698. [PMID: 31470787 DOI: 10.2174/1567205016666190830110152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/12/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022]
Abstract
Astrocytes are key cells for adequate brain formation and regulation of cerebral blood flow as well as for the maintenance of neuronal metabolism, neurotransmitter synthesis and exocytosis, and synaptic transmission. Many of these functions are intrinsically related to neurodegeneration, allowing refocusing on the role of astrocytes in physiological and neurodegenerative states. Indeed, emerging evidence in the field indicates that abnormalities in the astrocytic function are involved in the pathogenesis of multiple neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD) and Amyotrophic Lateral Sclerosis (ALS). In the present review, we highlight the physiological role of astrocytes in the CNS, including their communication with other cells in the brain. Furthermore, we discuss exciting findings and novel experimental approaches that elucidate the role of astrocytes in multiple neurological disorders.
Collapse
Affiliation(s)
- Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Angela Gomez-Arboledas
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Stefânia Forner
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Alessandra Cadete Martini
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States
| | - Antonia Gutierrez
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, University of Malaga, Malaga, Spain.,Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, CA 92697-4545, United States.,Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, United States
| |
Collapse
|
25
|
Type III intermediate filaments as targets and effectors of electrophiles and oxidants. Redox Biol 2020; 36:101582. [PMID: 32711378 PMCID: PMC7381704 DOI: 10.1016/j.redox.2020.101582] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/05/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Intermediate filaments (IFs) play key roles in cell mechanics, signaling and homeostasis. Their assembly and dynamics are finely regulated by posttranslational modifications. The type III IFs, vimentin, desmin, peripherin and glial fibrillary acidic protein (GFAP), are targets for diverse modifications by oxidants and electrophiles, for which their conserved cysteine residue emerges as a hot spot. Pathophysiological examples of these modifications include lipoxidation in cell senescence and rheumatoid arthritis, disulfide formation in cataracts and nitrosation in endothelial shear stress, although some oxidative modifications can also be detected under basal conditions. We previously proposed that cysteine residues of vimentin and GFAP act as sensors for oxidative and electrophilic stress, and as hinges influencing filament assembly. Accumulating evidence indicates that the structurally diverse cysteine modifications, either per se or in combination with other posttranslational modifications, elicit specific functional outcomes inducing distinct assemblies or network rearrangements, including filament stabilization, bundling or fragmentation. Cysteine-deficient mutants are protected from these alterations but show compromised cellular performance in network assembly and expansion, organelle positioning and aggresome formation, revealing the importance of this residue. Therefore, the high susceptibility to modification of the conserved cysteine of type III IFs and its cornerstone position in filament architecture sustains their role in redox sensing and integration of cellular responses. This has deep pathophysiological implications and supports the potential of this residue as a drug target. Type III intermediate filaments can be modified by many oxidants and electrophiles. Oxidative modifications of type III IFs occur in normal and pathological conditions. The conserved cysteine residue acts as a hub for redox/electrophilic modifications. Cysteine modifications elicit structure-dependent type III IF rearrangements. Type III intermediate filaments act as sensors for oxidative and electrophilic stress.
Collapse
|
26
|
Sung K, Jimenez-Sanchez M. Autophagy in Astrocytes and its Implications in Neurodegeneration. J Mol Biol 2020; 432:2605-2621. [PMID: 31931011 DOI: 10.1016/j.jmb.2019.12.041] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022]
Abstract
Autophagy is a major degradation pathway where double-membrane vesicles called autophagosomes deliver cytoplasmic content to the lysosome. Increasing evidence suggests that autophagy dysfunction contributes to the pathogenesis of neurodegenerative diseases. In addition, misfolded proteins that accumulate in these diseases and constitute a common pathological hallmark are substrates for autophagic degradation. Astrocytes, a major type of glial cells, are emerging as a critical component in most neurodegenerative diseases. This review will summarize the recent efforts to investigate the role that autophagy plays in astrocytes in the context of neurodegenerative diseases. While the field has mostly focused on the implications of autophagy in neurons, autophagy may also be involved in the clearance of disease-related proteins in astrocytes as well as in maintaining astrocyte function, which could impact the cell autonomous and non-cell autonomous contribution of astrocytes to neurodegeneration.
Collapse
Affiliation(s)
- Katherine Sung
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London, SE5 9RX, UK
| | - Maria Jimenez-Sanchez
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London, SE5 9RX, UK.
| |
Collapse
|
27
|
Abstract
Background Alexander disease is caused by dominantly acting mutations in glial fibrillary acidic protein (GFAP), the major intermediate filament of astrocytes in the central nervous system. Main body In addition to the sequence variants that represent the origin of disease, GFAP accumulation also takes place, together leading to a gain-of-function that has sometimes been referred to as “GFAP toxicity.” Whether the nature of GFAP toxicity in patients, who have mixtures of both mutant and normal protein, is the same as that produced by simple GFAP excess, is not yet clear. Conclusion The implications of these questions for the design of effective treatments are discussed.
Collapse
|
28
|
Sequeira J, Willson D, Marinello M. All in Your Mind? New-Onset Dysphagia in a Previously Healthy Adolescent Child. J Pediatr Intensive Care 2019; 9:135-138. [PMID: 32351769 DOI: 10.1055/s-0039-3401008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/03/2019] [Indexed: 10/25/2022] Open
Abstract
A healthy 11-year-old male develops fear of choking secondary to progressive dysphagia that began in early childhood. No organic cause is found, and the patient is diagnosed with psychiatric oral aversion. The child is eventually transferred to a psychiatric facility, where a month later he has a possible aspiration event and is transferred to the local emergency room for respiratory distress before being admitted to the intensive care unit. Workup is notable for abnormal findings on brain imaging, and the diagnosis of Alexander's disease is made. This case highlights the importance of complete history-taking and examinations in pediatric patients.
Collapse
Affiliation(s)
- Jake Sequeira
- VCU Department of Pediatric Critical Care, Richmond, Virginia, United States
| | - Douglas Willson
- VCU Department of Pediatric Critical Care, Richmond, Virginia, United States
| | - Mark Marinello
- VCU Department of Pediatric Critical Care, Richmond, Virginia, United States
| |
Collapse
|
29
|
Yasuda R, Nakano M, Yoshida T, Sato R, Adachi H, Tokuda Y, Mizuta I, Saito K, Matsuura J, Nakagawa M, Tashiro K, Mizuno T. Towards genomic database of Alexander disease to identify variations modifying disease phenotype. Sci Rep 2019; 9:14763. [PMID: 31611638 PMCID: PMC6791890 DOI: 10.1038/s41598-019-51390-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/01/2019] [Indexed: 01/13/2023] Open
Abstract
Alexander disease (AxD) is an extremely rare neurodegenerative disorder caused by glial fibrillary acidic protein (GFAP) gene mutations. Compared with the cerebral type, which is characterized by infantile onset, the bulbospinal type and intermediate form are associated with a late onset, spanning from juveniles to the elderly, and more diverse clinical spectrum, suggesting the existence of factors contributing to phenotypic diversity. To build a foundation for future genetic studies of this rare disease, we obtained genomic data by whole exome-sequencing (WES) and DNA microarray derived from thirty-one AxD patients with the bulbospinal type and intermediate form. Using this data, we aimed to identify genetic variations determining the age at onset (AAO) of AxD. As a result, WES- or microarray-based association studies between younger (<45 years; n = 13)- and older (≥45 years; n = 18)-onset patients considering the predicted GFAP-mutation pathogenicity identified no genome-wide significant variant. The candidate gene approach identified several variants likely correlated with AAO (p < 0.05): GAN, SLC1A2, CASP3, HDACs, and PI3K. Although we need to replicate the results using an independent population, this is the first step towards constructing a database, which may serve as an important tool to advance our understanding of AxD.
Collapse
Affiliation(s)
- Rei Yasuda
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masakazu Nakano
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomokatsu Yoshida
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Ryuichi Sato
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroko Adachi
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuichi Tokuda
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ikuko Mizuta
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kozo Saito
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun Matsuura
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masanori Nakagawa
- Department of Neurology, North Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Tashiro
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Toshiki Mizuno
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
30
|
Abstract
Leukodystrophies are genetically determined disorders affecting the white matter of the central nervous system. The combination of MRI pattern recognition and next-generation sequencing for the definition of novel disease entities has recently demonstrated that many leukodystrophies are due to the primary involvement and/or mutations in genes selectively expressed by cell types other than the oligodendrocytes, the myelin-forming cells in the brain. This has led to a new definition of leukodystrophies as genetic white matter disorders resulting from the involvement of any white matter structural component. As a result, the research has shifted its main focus from oligodendrocytes to other types of neuroglia. Astrocytes are the housekeeping cells of the nervous system, responsible for maintaining homeostasis and normal brain physiology and to orchestrate repair upon injury. Several lines of evidence show that astrocytic interactions with the other white matter cellular constituents play a primary pathophysiologic role in many leukodystrophies. These are thus now classified as astrocytopathies. This chapter addresses how the crosstalk between astrocytes, other glial cells, axons and non-neural cells are essential for the integrity and maintenance of the white matter in health. It also addresses the current knowledge of the cellular pathomechanisms of astrocytic leukodystrophies, and specifically Alexander disease, vanishing white matter, megalencephalic leukoencephalopathy with subcortical cysts and Aicardi-Goutière Syndrome.
Collapse
Affiliation(s)
- M S Jorge
- Department of Pathology, Free University Medical Centre, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Free University Medical Centre, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Klymkowsky MW. Filaments and phenotypes: cellular roles and orphan effects associated with mutations in cytoplasmic intermediate filament proteins. F1000Res 2019; 8. [PMID: 31602295 PMCID: PMC6774051 DOI: 10.12688/f1000research.19950.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2019] [Indexed: 12/11/2022] Open
Abstract
Cytoplasmic intermediate filaments (IFs) surround the nucleus and are often anchored at membrane sites to form effectively transcellular networks. Mutations in IF proteins (IFps) have revealed mechanical roles in epidermis, muscle, liver, and neurons. At the same time, there have been phenotypic surprises, illustrated by the ability to generate viable and fertile mice null for a number of IFp-encoding genes, including vimentin. Yet in humans, the vimentin ( VIM) gene displays a high probability of intolerance to loss-of-function mutations, indicating an essential role. A number of subtle and not so subtle IF-associated phenotypes have been identified, often linked to mechanical or metabolic stresses, some of which have been found to be ameliorated by the over-expression of molecular chaperones, suggesting that such phenotypes arise from what might be termed "orphan" effects as opposed to the absence of the IF network per se, an idea originally suggested by Toivola et al. and Pekny and Lane.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Molecular, Cellular & Developmental Biology, University of Colorado, Boulder, Boulder, CO, 80303, USA
| |
Collapse
|
32
|
Stadelmann C, Timmler S, Barrantes-Freer A, Simons M. Myelin in the Central Nervous System: Structure, Function, and Pathology. Physiol Rev 2019; 99:1381-1431. [PMID: 31066630 DOI: 10.1152/physrev.00031.2018] [Citation(s) in RCA: 394] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes generate multiple layers of myelin membrane around axons of the central nervous system to enable fast and efficient nerve conduction. Until recently, saltatory nerve conduction was considered the only purpose of myelin, but it is now clear that myelin has more functions. In fact, myelinating oligodendrocytes are embedded in a vast network of interconnected glial and neuronal cells, and increasing evidence supports an active role of oligodendrocytes within this assembly, for example, by providing metabolic support to neurons, by regulating ion and water homeostasis, and by adapting to activity-dependent neuronal signals. The molecular complexity governing these interactions requires an in-depth molecular understanding of how oligodendrocytes and axons interact and how they generate, maintain, and remodel their myelin sheaths. This review deals with the biology of myelin, the expanded relationship of myelin with its underlying axons and the neighboring cells, and its disturbances in various diseases such as multiple sclerosis, acute disseminated encephalomyelitis, and neuromyelitis optica spectrum disorders. Furthermore, we will highlight how specific interactions between astrocytes, oligodendrocytes, and microglia contribute to demyelination in hereditary white matter pathologies.
Collapse
Affiliation(s)
- Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Sebastian Timmler
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Alonso Barrantes-Freer
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Mikael Simons
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| |
Collapse
|
33
|
Heaven MR, Wilson L, Barnes S, Brenner M. Relative stabilities of wild-type and mutant glial fibrillary acidic protein in patients with Alexander disease. J Biol Chem 2019; 294:15604-15612. [PMID: 31484723 DOI: 10.1074/jbc.ra119.009777] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/24/2019] [Indexed: 01/13/2023] Open
Abstract
Alexander disease (AxD) is an often fatal astrogliopathy caused by dominant gain-of-function missense mutations in the glial fibrillary acidic protein (GFAP) gene. The mechanism by which the mutations produce the AxD phenotype is not known. However, the observation that features of AxD are displayed by mice that express elevated levels of GFAP from a human WT GFAP transgene has contributed to the notion that the mutations produce AxD by increasing accumulation of total GFAP above some toxic threshold rather than the mutant GFAP being inherently toxic. A possible mechanism for accumulation of GFAP in AxD patients is that the mutated GFAP variants are more stable than the WT, an attribution abetted by observations that GFAP complexes containing GFAP variants are more resistant to solvent extraction. Here we tested this hypothesis by determining the relative levels of WT and mutant GFAP in three individuals with AxD, each of whom carried a common but different GFAP mutation (R79C, R239H, or R416W). Mass spectrometry analysis identified a peptide specific to the mutant or WT GFAP in each patient, and we quantified this peptide by comparing its signal to that of an added [15N]GFAP standard. In all three individuals, the level of mutant GFAP was less than that of the WT. This finding suggests that AxD onset is due to an intrinsic toxicity of the mutant GFAP instead of it acting indirectly by being more stable than WT GFAP and thereby increasing the total GFAP level.
Collapse
Affiliation(s)
- Michael R Heaven
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294
| | - Landon Wilson
- Department of Pharmacology and Toxicology, Targeted Metabolomics and Proteomics Laboratory, University of Alabama, Birmingham, Alabama 35294
| | - Stephen Barnes
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294.,Department of Pharmacology and Toxicology, Targeted Metabolomics and Proteomics Laboratory, University of Alabama, Birmingham, Alabama 35294
| | - Michael Brenner
- Department of Neurobiology, University of Alabama, Birmingham, Alabama 35294
| |
Collapse
|
34
|
Abstract
Our understanding of astrocytes and their role in neurological diseases has increased considerably over the past two decades as the diverse roles of these cells have become recognized. Our evolving understanding of these cells suggests that they are more than support cells for neurons and that they play important roles in CNS homeostasis under normal conditions, in neuroprotection and in disease exacerbation. These multiple functions make them excellent candidates for targeted therapies to treat neurological disorders. New technological advances, including in vivo imaging, optogenetics and chemogenetics, have allowed us to examine astrocytic functions in ways that have uncovered new insights into the dynamic roles of these cells. Furthermore, the use of induced pluripotent stem cell-derived astrocytes from patients with a host of neurological disorders can help to tease out the contributions of astrocytes to human disease. In this Review, we explore some of the technological advances developed over the past decade that have aided our understanding of astrocyte function. We also highlight neurological disorders in which astrocyte function or dysfunction is believed to have a role in disease pathogenesis or propagation and discuss how the technological advances have been and could be used to study each of these diseases.
Collapse
|
35
|
Sosunov A, Olabarria M, Goldman JE. Alexander disease: an astrocytopathy that produces a leukodystrophy. Brain Pathol 2019; 28:388-398. [PMID: 29740945 DOI: 10.1111/bpa.12601] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/02/2018] [Indexed: 02/02/2023] Open
Abstract
Alexander Disease (AxD) is a degenerative disorder caused by mutations in the GFAP gene, which encodes the major intermediate filament of astrocytes. As other cells in the CNS do not express GFAP, AxD is a primary astrocyte disease. Astrocytes acquire a large number of pathological features, including changes in morphology, the loss or diminution of a number of critical astrocyte functions and the activation of cell stress and inflammatory pathways. AxD is also characterized by white matter degeneration, a pathology that has led it to be included in the "leukodystrophies." Furthermore, variable degrees of neuronal loss take place. Thus, the astrocyte pathology triggers alterations in other cell types. Here, we will review the neuropathology of AxD and discuss how a disease of astrocytes can lead to severe pathologies in non-astrocytic cells. Our knowledge of the pathophysiology of AxD will also lead to a better understanding of how astrocytes interact with other CNS cells and how astrocytes in the gliosis that accompanies many neurological disorders can damage the function and survival of other cells.
Collapse
Affiliation(s)
| | - Markel Olabarria
- Departments of Pathology and Cell Biology, Columbia University, New York, NY
| | - James E Goldman
- Departments of Pathology and Cell Biology, Columbia University, New York, NY
| |
Collapse
|
36
|
Kourtis N, Tavernarakis N. Small heat shock proteins and neurodegeneration: recent developments. Biomol Concepts 2018; 9:94-102. [PMID: 30133417 DOI: 10.1515/bmc-2018-0009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
AbstractMembers of the small heat shock protein (sHSP) family are molecular chaperones with a critical role in the maintenance of cellular homeostasis under unfavorable conditions. The chaperone properties of sHSPs prevent protein aggregation, and sHSP deregulation underlies the pathology of several diseases, including neurodegenerative disorders. Recent evidence suggests that the clientele of sHSPs is broad, and the mechanisms of sHSP-mediated neuroprotection diverse. Nonetheless, the crosstalk of sHSPs with the neurodegeneration-promoting signaling pathways remains poorly understood. Here, we survey recent findings on the role and regulation of sHSPs in neurodegenerative diseases.
Collapse
Affiliation(s)
- Nikos Kourtis
- Department of Pathology and Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, 70013, Crete, Greece.,Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, 71003, Crete, Greece
| |
Collapse
|
37
|
Tissue and cellular rigidity and mechanosensitive signaling activation in Alexander disease. Nat Commun 2018; 9:1899. [PMID: 29765022 PMCID: PMC5954157 DOI: 10.1038/s41467-018-04269-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 04/12/2018] [Indexed: 12/22/2022] Open
Abstract
Glial cells have increasingly been implicated as active participants in the pathogenesis of neurological diseases, but critical pathways and mechanisms controlling glial function and secondary non-cell autonomous neuronal injury remain incompletely defined. Here we use models of Alexander disease, a severe brain disorder caused by gain-of-function mutations in GFAP, to demonstrate that misregulation of GFAP leads to activation of a mechanosensitive signaling cascade characterized by activation of the Hippo pathway and consequent increased expression of A-type lamin. Importantly, we use genetics to verify a functional role for dysregulated mechanotransduction signaling in promoting behavioral abnormalities and non-cell autonomous neurodegeneration. Further, we take cell biological and biophysical approaches to suggest that brain tissue stiffness is increased in Alexander disease. Our findings implicate altered mechanotransduction signaling as a key pathological cascade driving neuronal dysfunction and neurodegeneration in Alexander disease, and possibly also in other brain disorders characterized by gliosis. Alexander disease is a rare neurodegeneration caused by mutations in a glial gene GFAP. Here, Wang and colleagues show in animal models of Alexander disease that GFAP mutant brain and cells have greater tissue and cellular stiffness and greater activation of mechanosensitive signaling cascade.
Collapse
|
38
|
Perez-Nievas BG, Serrano-Pozo A. Deciphering the Astrocyte Reaction in Alzheimer's Disease. Front Aging Neurosci 2018; 10:114. [PMID: 29922147 PMCID: PMC5996928 DOI: 10.3389/fnagi.2018.00114] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/03/2018] [Indexed: 12/24/2022] Open
Abstract
Reactive astrocytes were identified as a component of senile amyloid plaques in the cortex of Alzheimer's disease (AD) patients several decades ago. However, their role in AD pathophysiology has remained elusive ever since, in part owing to the extrapolation of the literature from primary astrocyte cultures and acute brain injury models to a chronic neurodegenerative scenario. Recent accumulating evidence supports the idea that reactive astrocytes in AD acquire neurotoxic properties, likely due to both a gain of toxic function and a loss of their neurotrophic effects. However, the diversity and complexity of this glial cell is only beginning to be unveiled, anticipating that astrocyte reaction might be heterogeneous as well. Herein we review the evidence from mouse models of AD and human neuropathological studies and attempt to decipher the main conundrums that astrocytes pose to our understanding of AD development and progression. We discuss the morphological features that characterize astrocyte reaction in the AD brain, the consequences of astrocyte reaction for both astrocyte biology and AD pathological hallmarks, and the molecular pathways that have been implicated in this reaction.
Collapse
Affiliation(s)
| | - Alberto Serrano-Pozo
- Alzheimer's Research Unit, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
39
|
Zhu Z, Reiser G. The small heat shock proteins, especially HspB4 and HspB5 are promising protectants in neurodegenerative diseases. Neurochem Int 2018; 115:69-79. [PMID: 29425965 DOI: 10.1016/j.neuint.2018.02.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/24/2018] [Accepted: 02/05/2018] [Indexed: 12/13/2022]
Abstract
Small heat shock proteins (sHsps) are a group of proteins with molecular mass between 12 and 43 kDa. Currently, 11 members of this family have been classified, namely HspB1 to HspB11. HspB1, HspB2, HspB5, HspB6, HspB7, and HspB8, which are expressed in brain have been observed to be related to the pathology of neurodegenerative diseases, including Parkinson's, Alzheimer's, Alexander's disease, multiple sclerosis, and human immunodeficiency virus-associated dementia. Specifically, sHsps interact with misfolding and damaging protein aggregates, like Glial fibrillary acidic protein in AxD, β-amyloid peptides aggregates in Alzheimer's disease, Superoxide dismutase 1 in Amyotrophic lateral sclerosis and cytosine-adenine-guanine/polyglutamine (CAG/PolyQ) in Huntington's disease, Spinocerebellar ataxia type 3, Spinal-bulbar muscular atrophy, to reduce the toxicity or increase the clearance of these protein aggregates. The degree of HspB4 expression in brain is still debated. For neuroprotective mechanisms, sHsps attenuate mitochondrial dysfunctions, reduce accumulation of misfolded proteins, block oxidative/nitrosative stress, and minimize neuronal apoptosis and neuroinflammation, which are molecular mechanisms commonly accepted to mirror the progression and development of neurodegenerative diseases. The increasing incidence of the neurodegenerative diseases enhanced search for effective approaches to rescue neural tissue from degeneration with minimal side effects. sHsps have been found to exert neuroprotective functions. HspB5 has been emphasized to reduce the paralysis in a mouse model of experimental autoimmune encephalomyelitis, providing a therapeutic basis for the disease. In this review, we discuss the current understanding of the properties and the mechanisms of protection orchestrated by sHsps in the nervous system, highlighting the promising therapeutic role of sHsps in neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhihui Zhu
- Institut für Inflammation und Neurodegeneration (Neurobiochemie), Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Leipziger Straße 44, 39120 Magdeburg, Germany; College of Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Georg Reiser
- Institut für Inflammation und Neurodegeneration (Neurobiochemie), Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Leipziger Straße 44, 39120 Magdeburg, Germany.
| |
Collapse
|
40
|
Saito K, Shigetomi E, Yasuda R, Sato R, Nakano M, Tashiro K, Tanaka KF, Ikenaka K, Mikoshiba K, Mizuta I, Yoshida T, Nakagawa M, Mizuno T, Koizumi S. Aberrant astrocyte Ca 2+ signals "AxCa signals" exacerbate pathological alterations in an Alexander disease model. Glia 2018; 66:1053-1067. [PMID: 29383757 DOI: 10.1002/glia.23300] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 12/12/2017] [Accepted: 01/10/2018] [Indexed: 12/21/2022]
Abstract
Alexander disease (AxD) is a rare neurodegenerative disorder caused by gain of function mutations in the glial fibrillary acidic protein (GFAP) gene. Accumulation of GFAP proteins and formation of Rosenthal fibers (RFs) in astrocytes are hallmarks of AxD. However, malfunction of astrocytes in the AxD brain is poorly understood. Here, we show aberrant Ca2+ responses in astrocytes as playing a causative role in AxD. Transcriptome analysis of astrocytes from a model of AxD showed age-dependent upregulation of GFAP, several markers for neurotoxic reactive astrocytes, and downregulation of Ca2+ homeostasis molecules. In situ AxD model astrocytes produced aberrant extra-large Ca2+ signals "AxCa signals", which increased with age, correlated with GFAP upregulation, and were dependent on stored Ca2+ . Inhibition of AxCa signals by deletion of inositol 1,4,5-trisphosphate type 2 receptors (IP3R2) ameliorated AxD pathogenesis. Taken together, AxCa signals in the model astrocytes would contribute to AxD pathogenesis.
Collapse
Affiliation(s)
- Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Kofu, Yamanashi Prefecture, 400-8510, Japan.,Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Kofu, Yamanashi Prefecture, 400-8510, Japan
| | - Rei Yasuda
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryuichi Sato
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masakazu Nakano
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Tashiro
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenji F Tanaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Wako, Japan
| | - Ikuko Mizuta
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomokatsu Yoshida
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masanori Nakagawa
- Department of Neurology, North Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiki Mizuno
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Kofu, Yamanashi Prefecture, 400-8510, Japan
| |
Collapse
|
41
|
Hagemann TL, Powers B, Mazur C, Kim A, Wheeler S, Hung G, Swayze E, Messing A. Antisense suppression of glial fibrillary acidic protein as a treatment for Alexander disease. Ann Neurol 2018; 83:27-39. [PMID: 29226998 DOI: 10.1002/ana.25118] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/24/2017] [Accepted: 12/06/2017] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Alexander disease is a fatal leukodystrophy caused by autosomal dominant gain-of-function mutations in the gene for glial fibrillary acidic protein (GFAP), an intermediate filament protein primarily expressed in astrocytes of the central nervous system. A key feature of pathogenesis is overexpression and accumulation of GFAP, with formation of characteristic cytoplasmic aggregates known as Rosenthal fibers. Here we investigate whether suppressing GFAP with antisense oligonucleotides could provide a therapeutic strategy for treating Alexander disease. METHODS In this study, we use GFAP mutant mouse models of Alexander disease to test the efficacy of antisense suppression and evaluate the effects on molecular and cellular phenotypes and non-cell-autonomous toxicity. Antisense oligonucleotides were designed to target the murine Gfap transcript, and screened using primary mouse cortical cultures. Lead oligonucleotides were then tested for their ability to reduce GFAP transcripts and protein, first in wild-type mice with normal levels of GFAP, and then in adult mutant mice with established pathology and elevated levels of GFAP. RESULTS Nearly complete and long-lasting elimination of GFAP occurred in brain and spinal cord following single bolus intracerebroventricular injections, with a striking reversal of Rosenthal fibers and downstream markers of microglial and other stress-related responses. GFAP protein was also cleared from cerebrospinal fluid, demonstrating its potential utility as a biomarker in future clinical applications. Finally, treatment led to improved body condition and rescue of hippocampal neurogenesis. INTERPRETATION These results demonstrate the efficacy of antisense suppression for an astrocyte target, and provide a compelling therapeutic approach for Alexander disease. Ann Neurol 2018;83:27-39.
Collapse
Affiliation(s)
| | | | | | | | - Steven Wheeler
- Waisman Center, University of Wisconsin-Madison, Madison, WI
| | | | | | - Albee Messing
- Waisman Center, University of Wisconsin-Madison, Madison, WI.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
42
|
van der Knaap MS, Bugiani M. Leukodystrophies: a proposed classification system based on pathological changes and pathogenetic mechanisms. Acta Neuropathol 2017; 134:351-382. [PMID: 28638987 PMCID: PMC5563342 DOI: 10.1007/s00401-017-1739-1] [Citation(s) in RCA: 252] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/06/2017] [Accepted: 06/06/2017] [Indexed: 12/29/2022]
Abstract
Leukodystrophies are genetically determined disorders characterized by the selective involvement of the central nervous system white matter. Onset may be at any age, from prenatal life to senescence. Many leukodystrophies are degenerative in nature, but some only impair white matter function. The clinical course is mostly progressive, but may also be static or even improving with time. Progressive leukodystrophies are often fatal, and no curative treatment is known. The last decade has witnessed a tremendous increase in the number of defined leukodystrophies also owing to a diagnostic approach combining magnetic resonance imaging pattern recognition and next generation sequencing. Knowledge on white matter physiology and pathology has also dramatically built up. This led to the recognition that only few leukodystrophies are due to mutations in myelin- or oligodendrocyte-specific genes, and many are rather caused by defects in other white matter structural components, including astrocytes, microglia, axons and blood vessels. We here propose a novel classification of leukodystrophies that takes into account the primary involvement of any white matter component. Categories in this classification are the myelin disorders due to a primary defect in oligodendrocytes or myelin (hypomyelinating and demyelinating leukodystrophies, leukodystrophies with myelin vacuolization); astrocytopathies; leuko-axonopathies; microgliopathies; and leuko-vasculopathies. Following this classification, we illustrate the neuropathology and disease mechanisms of some leukodystrophies taken as example for each category. Some leukodystrophies fall into more than one category. Given the complex molecular and cellular interplay underlying white matter pathology, recognition of the cellular pathology behind a disease becomes crucial in addressing possible treatment strategies.
Collapse
Affiliation(s)
- Marjo S van der Knaap
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Functional Genomics, Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands.
- Department of Pathology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Olabarria M, Goldman JE. Disorders of Astrocytes: Alexander Disease as a Model. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 12:131-152. [PMID: 28135564 DOI: 10.1146/annurev-pathol-052016-100218] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Astrocytes undergo important phenotypic changes in many neurological disorders, including strokes, trauma, inflammatory diseases, infectious diseases, and neurodegenerative diseases. We have been studying the astrocytes of Alexander disease (AxD), which is caused by heterozygous mutations in the GFAP gene, which is the gene that encodes the major astrocyte intermediate filament protein. AxD is a primary astrocyte disease because GFAP expression is specific to astrocytes in the central nervous system (CNS). The accumulation of extremely large amounts of GFAP causes many molecular changes in astrocytes, including proteasome inhibition, stress kinase activation, mechanistic target of rapamycin (mTOR) activation, loss of glutamate and potassium buffering capacity, loss of astrocyte coupling, and changes in cell morphology. Many of these changes appear to be common to astrocyte reactions in other neurological disorders. Using AxD to illuminate common mechanisms, we discuss the molecular pathology of AxD astrocytes and compare that to astrocyte pathology in other disorders.
Collapse
Affiliation(s)
- Markel Olabarria
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; ,
| | - James E Goldman
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032; ,
| |
Collapse
|
44
|
Lin NH, Messing A, Perng MD. Characterization of a panel of monoclonal antibodies recognizing specific epitopes on GFAP. PLoS One 2017; 12:e0180694. [PMID: 28700643 PMCID: PMC5503259 DOI: 10.1371/journal.pone.0180694] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 06/20/2017] [Indexed: 11/19/2022] Open
Abstract
Alexander disease (AxD) is a neurodegenerative disease caused by heterozygous mutations in the GFAP gene, which encodes the major intermediate filament protein of astrocytes. This disease is characterized by the accumulation of cytoplasmic protein aggregates, known as Rosenthal fibers. Antibodies specific to GFAP could provide invaluable tools to facilitate studies of the normal biology of GFAP and to elucidate the pathologic role of this IF protein in disease. While a large number of antibodies to GFAP are available, few if any of them have defined epitopes. Here we described the characterization of a panel of commonly used anti-GFAP antibodies, which recognized epitopes at regions extending across the rod domain of GFAP. We show that all of the antibodies are useful for immunoblotting and immunostaining, and identify a subset that preferentially recognized human GFAP. Using these antibodies, we demonstrate the presence of biochemically modified forms of GFAP in brains of human AxD patients and mouse AxD models. These data suggest that this panel of anti-GFAP antibodies will be useful for studies of animal and cell-based models of AxD and related diseases in which cytoskeletal defects associated with GFAP modifications occur.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Albee Messing
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ming-Der Perng
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
- * E-mail:
| |
Collapse
|
45
|
Gómez-Pinedo U, Sirerol-Piquer MS, Durán-Moreno M, García-Verdugo JM, Matias-Guiu J. Alexander Disease Mutations Produce Cells with Coexpression of Glial Fibrillary Acidic Protein and NG2 in Neurosphere Cultures and Inhibit Differentiation into Mature Oligodendrocytes. Front Neurol 2017; 8:255. [PMID: 28634469 PMCID: PMC5459916 DOI: 10.3389/fneur.2017.00255] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 05/22/2017] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Alexander disease (AxD) is a rare disease caused by mutations in the gene encoding glial fibrillary acidic protein (GFAP). The disease is characterized by presence of GFAP aggregates in the cytoplasm of astrocytes and loss of myelin. OBJECTIVES Determine the effect of AxD-related mutations on adult neurogenesis. METHODS We transfected different types of mutant GFAP into neurospheres using the nucleofection technique. RESULTS We find that mutations may cause coexpression of GFAP and NG2 in neurosphere cultures, which would inhibit the differentiation of precursors into oligodendrocytes and thus explain the myelin loss occurring in the disease. Transfection produces cells that differentiate into new cells marked simultaneously by GFAP and NG2 and whose percentage increased over days of differentiation. Increased expression of GFAP is due to a protein with an anomalous structure that forms aggregates throughout the cytoplasm of new cells. These cells display down-expression of vimentin and nestin. Up-expression of cathepsin D and caspase-3 in the first days of differentiation suggest that apoptosis as a lysosomal response may be at work. HSP27, a protein found in Rosenthal bodies, is expressed less at the beginning of the process although its presence increases in later stages. CONCLUSION Our findings seem to suggest that the mechanism of development of AxD may not be due to a function gain due to increase of GFAP, but to failure in the differentiation process may occur at the stage in which precursor cells transform into oligodendrocytes, and that possibility may provide the best explanation for the clinical and radiological images described in AxD.
Collapse
Affiliation(s)
- Ulises Gómez-Pinedo
- Neurobiology Laboratory, Neuroscience Institute, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Maria Salomé Sirerol-Piquer
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biologia Evolutiva, Universidad de Valencia, Valencia, Spain
| | - María Durán-Moreno
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biologia Evolutiva, Universidad de Valencia, Valencia, Spain
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biologia Evolutiva, Universidad de Valencia, Valencia, Spain
| | - Jorge Matias-Guiu
- Neurobiology Laboratory, Neuroscience Institute, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
46
|
Sosunov AA, McKhann GM, Goldman JE. The origin of Rosenthal fibers and their contributions to astrocyte pathology in Alexander disease. Acta Neuropathol Commun 2017; 5:27. [PMID: 28359321 PMCID: PMC5374671 DOI: 10.1186/s40478-017-0425-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/08/2017] [Indexed: 11/27/2022] Open
Abstract
Rosenthal fibers (RFs) are cytoplasmic, proteinaceous aggregates. They are the pathognomonic feature of the astrocyte pathology in Alexander Disease (AxD), a neurodegenerative disorder caused by heterozygous mutations in the GFAP gene, encoding glial fibrillary acidic protein (GFAP). Although RFs have been known for many years their origin and significance remain elusive issues. We have used mouse models of AxD based on the overexpression of human GFAP (transgenic, TG) and a point mutation in mouse GFAP (knock-in, KI) to examine the formation of RFs and to find astrocyte changes that correlate with the appearance of RFs. We found RFs of various sizes and shapes. The smallest ones appear as granular depositions on intermediate filaments. These contain GFAP and the small heat shock protein, alphaB-crystallin. Their aggregation appears to give rise to large RFs. The appearance of new RFs and the growth of previously formed RFs occur over time. We determined that DAPI is a reliable marker of RFs and in parallel with Fluoro-Jade B (FJB) staining defined a high variability in the appearance of RFs, even in neighboring astrocytes. Although many astrocytes in AxD with increased levels of GFAP and with or without RFs change their phenotype, only some cells with large numbers of RFs show a profound reconstruction of cellular processes, with a loss of fine distal processes and the appearance of large, lobulated nuclei, likely due to arrested mitosis. We conclude that 1) RFs appear to originate as small, osmiophilic masses containing both GFAP and alphaB-crystallin deposited on bundles of intermediate filaments. 2) RFs continue to form within AxD astrocytes over time. 3) DAPI is a reliable marker for RFs and can be used with immunolabeling. 4) RFs appear to interfere with the successful completion of astrocyte mitosis and cell division.
Collapse
|
47
|
Moody LR, Barrett-Wilt GA, Sussman MR, Messing A. Glial fibrillary acidic protein exhibits altered turnover kinetics in a mouse model of Alexander disease. J Biol Chem 2017; 292:5814-5824. [PMID: 28223355 DOI: 10.1074/jbc.m116.772020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/15/2017] [Indexed: 12/27/2022] Open
Abstract
Mutations in the astrocyte-specific intermediate filament glial fibrillary acidic protein (GFAP) lead to the rare and fatal disorder, Alexander disease (AxD). A prominent feature of the disease is aberrant accumulation of GFAP. It has been proposed that this accumulation occurs because of an increase in gene transcription coupled with impaired proteasomal degradation, yet this hypothesis remains untested. We therefore sought to directly investigate GFAP turnover in a mouse model of AxD that is heterozygous for a disease-causing point mutation (GfapR236H/+) (and thus expresses both wild-type and mutant protein). Stable isotope labeling by amino acids in cell culture, using primary cortical astrocytes, indicated that the in vitro half-lives of total GFAP in astrocytes from wild-type and mutant mice were similar at ∼3-4 days. Surprisingly, results obtained with stable isotope labeling of mammals revealed that, in vivo, the half-life of GFAP in mutant mice (15.4 ± 0.5 days) was much shorter than that in wild-type mice (27.5 ± 1.6 days). These unexpected in vivo data are most consistent with a model in which synthesis and degradation are both increased. Our work reveals that an AxD-causing mutation alters GFAP turnover kinetics in vivo and provides an essential foundation for future studies aimed at preventing or reducing the accumulation of GFAP. In particular, these data suggest that elimination of GFAP might be possible and occurs more quickly than previously surmised.
Collapse
Affiliation(s)
| | | | | | - Albee Messing
- From the Waisman Center, .,Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|
48
|
Lin NH, Huang YS, Opal P, Goldman RD, Messing A, Perng MD. The role of gigaxonin in the degradation of the glial-specific intermediate filament protein GFAP. Mol Biol Cell 2016; 27:3980-3990. [PMID: 27798231 PMCID: PMC5156539 DOI: 10.1091/mbc.e16-06-0362] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 10/03/2016] [Accepted: 10/19/2016] [Indexed: 01/04/2023] Open
Abstract
Alexander disease (AxD) is a primary genetic disorder of astrocytes caused by dominant mutations in the gene encoding the intermediate filament (IF) protein GFAP. This disease is characterized by excessive accumulation of GFAP, known as Rosenthal fibers, within astrocytes. Abnormal GFAP aggregation also occurs in giant axon neuropathy (GAN), which is caused by recessive mutations in the gene encoding gigaxonin. Given that one of the functions of gigaxonin is to facilitate proteasomal degradation of several IF proteins, we sought to determine whether gigaxonin is involved in the degradation of GFAP. Using a lentiviral transduction system, we demonstrated that gigaxonin levels influence the degradation of GFAP in primary astrocytes and in cell lines that express this IF protein. Gigaxonin was similarly involved in the degradation of some but not all AxD-associated GFAP mutants. In addition, gigaxonin directly bound to GFAP, and inhibition of proteasome reversed the clearance of GFAP in cells achieved by overexpressing gigaxonin. These studies identify gigaxonin as an important factor that targets GFAP for degradation through the proteasome pathway. Our findings provide a critical foundation for future studies aimed at reducing or reversing pathological accumulation of GFAP as a potential therapeutic strategy for AxD and related diseases.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yu-Shan Huang
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Puneet Opal
- Davee Department of Neurology, Northwestern University, Chicago, IL 60611.,Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Albee Messing
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705
| | - Ming-Der Perng
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
49
|
Cortese K, Daga A, Monticone M, Tavella S, Stefanelli A, Aiello C, Bisio A, Bellese G, Castagnola P. Carnosic acid induces proteasomal degradation of Cyclin B1, RB and SOX2 along with cell growth arrest and apoptosis in GBM cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:679-685. [PMID: 27235706 DOI: 10.1016/j.phymed.2016.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 03/14/2016] [Accepted: 03/19/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND Carnosic acid (CA) is a diterpenoid found in Rosmarinus officinalis L. and Salvia officinalis L. as well as in many other Lamiaceae. This compound is reported to have antioxidant and antimicrobial properties. In addition, a number of reports showed that CA has a cytotoxic activity toward several cancer cell lines. PURPOSE The aim of this study was to establish whether CA has any specific antiproliferative effect toward human glioblastoma (GBM) cells and to analyze the molecular mechanisms involved. METHODS We evaluated cell survival by MTT assay, apoptosis and DNA content by flow cytometry, protein expression and phosphorylation by immunoblot analyses. RESULTS Our results showed that CA inhibited cell survival on both normal astrocytes and GBM cells. In GBM cells, in particular, CA caused an early G2 block, a reduction in the percentage of cells expressing Ki67, an enhanced expression of p21(WAF) and induced apoptosis. Furthermore, we showed that CA promoted proteasomal degradation of several substrate proteins, including Cyclin B1, retinoblastoma (RB), SOX2, and glial fibrillary acid protein (GFAP), whereas MYC levels were not modified. In addition, CA dramatically reduced the activity of CDKs. CONCLUSION In conclusion, our findings strongly suggest that CA promotes a profound deregulation of cell cycle control and reduces the survival of GBM cells via proteasome-mediated degradation of Cyclin B1, RB and SOX2.
Collapse
Affiliation(s)
- Katia Cortese
- DIMES, Dipartimento di Medicina Sperimentale, Anatomia Umana, Università di Genova, Via de Toni 14, 16132 Genova, Italy
| | - Antonio Daga
- IRCCS AOU - San Martino - IST, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | | | - Sara Tavella
- DIMES, Dipartimento di Medicina Sperimentale, Anatomia Umana, Università di Genova, Via de Toni 14, 16132 Genova, Italy; IRCCS AOU - San Martino - IST, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Alessia Stefanelli
- IRCCS AOU - San Martino - IST, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Cinzia Aiello
- IRCCS AOU - San Martino - IST, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Angela Bisio
- Dip. Farmacia, Università di Genova, Via Brigata Salerno 13, 16147 Genova, Italy
| | - Grazia Bellese
- DIMES, Dipartimento di Medicina Sperimentale, Anatomia Umana, Università di Genova, Via de Toni 14, 16132 Genova, Italy
| | - Patrizio Castagnola
- IRCCS AOU - San Martino - IST, Largo Rosanna Benzi, 10, 16132 Genova, Italy.
| |
Collapse
|
50
|
Molecular Pathophysiology of Fragile X-Associated Tremor/Ataxia Syndrome and Perspectives for Drug Development. THE CEREBELLUM 2016; 15:599-610. [DOI: 10.1007/s12311-016-0800-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|