1
|
Xiang H. The interplay between α-synuclein aggregation and necroptosis in Parkinson's disease: a spatiotemporal perspective. Front Neurosci 2025; 19:1567445. [PMID: 40264913 PMCID: PMC12011736 DOI: 10.3389/fnins.2025.1567445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the death of dopaminergic neurons and the aggregation of alpha-synuclein (α-Syn). It presents with prominent motor symptoms, and by the time of diagnosis, a significant number of neurons have already been lost. Current medications can only alleviate symptoms but cannot halt disease progression. Studies have confirmed that both dopaminergic neuronal loss and α-Syn aggregation are associated with necroptosis mechanisms. Necroptosis, a regulated form of cell death, has been recognized as an underexplored hotspot in PD pathogenesis research. In this review, we propose a spatiotemporal model of PD progression, highlighting the interactions between α-Syn aggregation, mitochondrial dysfunction, oxidative stress, neuroinflammation and necroptosis. These processes not only drive motor symptoms but also contribute to early non-motor symptoms, offering insights into potential diagnostic markers. Finally, we touch upon the therapeutic potential of necroptosis inhibition in enhancing current PD treatments, such as L-Dopa. This review aims to provide a new perspective on the pathogenesis of PD and to identify avenues for the development of more effective therapeutic strategies.
Collapse
Affiliation(s)
- Haoran Xiang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
- Department of Neurology, Yichang Central People’s Hospital, Yichang, Hubei, China
| |
Collapse
|
2
|
Yang J, Tang C. Causal relationship between imaging-derived phenotypes and neurodegenerative diseases: a Mendelian randomization study. Mamm Genome 2024; 35:711-723. [PMID: 39180568 DOI: 10.1007/s00335-024-10065-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024]
Abstract
Neurodegenerative diseases are incurable conditions that lead to gradual and progressive deterioration of brain function in patients. With the aging population, the prevalence of these diseases is expected to increase, posing a significant economic burden on society. Imaging techniques play a crucial role in the diagnosis and monitoring of neurodegenerative diseases. This study utilized a two-sample Mendelian randomization (MR) analysis to assess the causal relationship between different imaging-derived phenotypes (IDP) in the brain and neurodegenerative diseases. Multiple MR methods were employed to minimize bias and obtain reliable estimates of the potential causal relationship between the variable exposures of interest and the outcomes. The study found potential causal relationships between different IDPs and Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and frontotemporal dementia (FTD). Specifically, the study identified potential causal relationships between 2 different types of IDPs and AD, 8 different types of IDPs and PD, 11 different types of imaging-derived phenotypes and ALS, 1 type of IDP and MS, and 1 type of IDP and FTD. This study provides new insights for the prevention, diagnosis, and treatment of neurodegenerative diseases, offering important clues for understanding the pathogenesis of these diseases and developing relevant intervention strategies.
Collapse
Affiliation(s)
- Jiaxin Yang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Chao Tang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China.
- School of Clinical Medicine, Guizhou Medical University, No.28, Guiyi Street, Yunyan District, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
3
|
Xu Z, He S, Begum MM, Han X. Myelin Lipid Alterations in Neurodegenerative Diseases: Landscape and Pathogenic Implications. Antioxid Redox Signal 2024; 41:1073-1099. [PMID: 39575748 PMCID: PMC11971557 DOI: 10.1089/ars.2024.0676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024]
Abstract
Significance: Lipids, which constitute the highest portion (over 50%) of brain dry mass, are crucial for brain integrity, energy homeostasis, and signaling regulation. Emerging evidence revealed that lipid profile alterations and abnormal lipid metabolism occur during normal aging and in different forms of neurodegenerative diseases. Moreover, increasing genome-wide association studies have validated new targets on lipid-associated pathways involved in disease development. Myelin, the protective sheath surrounding axons, is crucial for efficient neural signaling transduction. As the primary site enriched with lipids, impairments of myelin are increasingly recognized as playing significant and complex roles in various neurodegenerative diseases, beyond simply being secondary effects of neuronal loss. Recent Advances: With advances in the lipidomics field, myelin lipid alterations and their roles in contributing to or reflecting the progression of diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and others, have recently caught great attention. Critical Issues: This review summarizes recent findings of myelin lipid alterations in the five most common neurodegenerative diseases and discusses their implications in disease pathogenesis. Future Directions: By highlighting myelin lipid abnormalities in neurodegenerative diseases, this review aims to encourage further research focused on lipids and the development of new lipid-oriented therapeutic approaches in this area. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ziying Xu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Sijia He
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Mst Marium Begum
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
- Department of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
4
|
Sánchez de la Torre A, Ezquerro-Herce S, Huerga-Gómez A, Sánchez-Martín E, Chara JC, Matute C, Monory K, Mato S, Lutz B, Guzmán M, Aguado T, Palazuelos J. CB 1 receptors in NG2 cells mediate cannabinoid-evoked functional myelin regeneration. Prog Neurobiol 2024; 243:102683. [PMID: 39528076 DOI: 10.1016/j.pneurobio.2024.102683] [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: 07/02/2024] [Revised: 09/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Defects in myelin homeostasis have been reported in many neuropathological conditions. Cannabinoid compounds have been shown to efficiently promote myelin regeneration in animal models of demyelination. However, it is still unknown whether this action relies mostly on a cell autonomous effect on oligodendroglial-lineage-NG2 cells. By using conditional genetic mouse models, here we found that cannabinoid CB1 receptors located on NG2 cells are required for oligodendroglial differentiation and myelin regeneration after demyelination. Selective CB1 receptor gene depletion in NG2 cells following toxin-induced demyelination disrupted oligodendrocyte regeneration and functional remyelination and exacerbated axonal damage. These deficits were rescued by pharmacological blockade of the RhoA/ROCK/Cofilin pathway. Conversely, tetrahydrocannabinol administration promoted oligodendrocyte regeneration and functional remyelination in wild-type but not Ng2-CB1-deficient mice. Overall, this study identifies CB1 receptors as essential modulators of remyelination and support the therapeutic potential of cannabinoids for promoting remyelination in neurological disorders.
Collapse
Affiliation(s)
- Aníbal Sánchez de la Torre
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain; Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid 28040, Spain
| | - Sara Ezquerro-Herce
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain; Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid 28040, Spain
| | - Alba Huerga-Gómez
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain; Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid 28040, Spain
| | - Ester Sánchez-Martín
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa 48940, Spain; Achucarro Basque Center for Neuroscience, Leioa 48940, Spain; IIS Biobizkaia, Barakaldo 48903, Spain
| | - Juan Carlos Chara
- Achucarro Basque Center for Neuroscience, Leioa 48940, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid 28049, Spain
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa 48940, Spain; Achucarro Basque Center for Neuroscience, Leioa 48940, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid 28049, Spain
| | - Krisztina Monory
- Institute of Physiological Chemistry, University Medical Center Mainz, Mainz 55128, Germany
| | - Susana Mato
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa 48940, Spain; Achucarro Basque Center for Neuroscience, Leioa 48940, Spain; Institute of Physiological Chemistry, University Medical Center Mainz, Mainz 55128, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center Mainz, Mainz 55128, Germany
| | - Manuel Guzmán
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain; Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid 28040, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid 28049, Spain
| | - Tania Aguado
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain; Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid 28040, Spain.
| | - Javier Palazuelos
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain; Department of Biochemistry and Molecular Biology, Complutense University, Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid 28040, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid 28049, Spain.
| |
Collapse
|
5
|
Tangavelou K, Bhaskar K. The Mechanistic Link Between Tau-Driven Proteotoxic Stress and Cellular Senescence in Alzheimer's Disease. Int J Mol Sci 2024; 25:12335. [PMID: 39596399 PMCID: PMC11595124 DOI: 10.3390/ijms252212335] [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: 10/11/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
In Alzheimer's disease (AD), tau dissociates from microtubules (MTs) due to hyperphosphorylation and misfolding. It is degraded by various mechanisms, including the 20S proteasome, chaperone-mediated autophagy (CMA), 26S proteasome, macroautophagy, and aggrephagy. Neurofibrillary tangles (NFTs) form upon the impairment of aggrephagy, and eventually, the ubiquitin chaperone valosin-containing protein (VCP) and heat shock 70 kDa protein (HSP70) are recruited to the sites of NFTs for the extraction of tau for the ubiquitin-proteasome system (UPS)-mediated degradation. However, the impairment of tau degradation in neurons allows tau to be secreted into the extracellular space. Secreted tau can be monomers, oligomers, and paired helical filaments (PHFs), which are seeding competent pathological tau that can be endocytosed/phagocytosed by healthy neurons, microglia, astrocytes, oligodendrocyte progenitor cells (OPCs), and oligodendrocytes, often causing proteotoxic stress and eventually triggers senescence. Senescent cells secrete various senescence-associated secretory phenotype (SASP) factors, which trigger cellular atrophy, causing decreased brain volume in human AD. However, the molecular mechanisms of proteotoxic stress and cellular senescence are not entirely understood and are an emerging area of research. Therefore, this comprehensive review summarizes pertinent studies that provided evidence for the sequential tau degradation, failure, and the mechanistic link between tau-driven proteotoxic stress and cellular senescence in AD.
Collapse
Affiliation(s)
- Karthikeyan Tangavelou
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
6
|
Helgudóttir SS, Mørkholt AS, Lichota J, Bruun-Nyzell P, Andersen MC, Kristensen NMJ, Johansen AK, Zinn MR, Jensdóttir HM, Nieland JDV. Rethinking neurodegenerative diseases: neurometabolic concept linking lipid oxidation to diseases in the central nervous system. Neural Regen Res 2024; 19:1437-1445. [PMID: 38051885 PMCID: PMC10883494 DOI: 10.4103/1673-5374.387965] [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: 05/31/2023] [Revised: 08/31/2023] [Accepted: 09/21/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Currently, there is a lack of effective medicines capable of halting or reversing the progression of neurodegenerative disorders, including amyotrophic lateral sclerosis, Parkinson's disease, multiple sclerosis, or Alzheimer's disease. Given the unmet medical need, it is necessary to reevaluate the existing paradigms of how to target these diseases. When considering neurodegenerative diseases from a systemic neurometabolic perspective, it becomes possible to explain the shared pathological features. This innovative approach presented in this paper draws upon extensive research conducted by the authors and researchers worldwide. In this review, we highlight the importance of metabolic mitochondrial dysfunction in the context of neurodegenerative diseases. We provide an overview of the risk factors associated with developing neurodegenerative disorders, including genetic, epigenetic, and environmental factors. Additionally, we examine pathological mechanisms implicated in these diseases such as oxidative stress, accumulation of misfolded proteins, inflammation, demyelination, death of neurons, insulin resistance, dysbiosis, and neurotransmitter disturbances. Finally, we outline a proposal for the restoration of mitochondrial metabolism, a crucial aspect that may hold the key to facilitating curative therapeutic interventions for neurodegenerative disorders in forthcoming advancements.
Collapse
Affiliation(s)
| | | | - Jacek Lichota
- Molecular Pharmacology Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | - Mads Christian Andersen
- Molecular Pharmacology Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Nanna Marie Juhl Kristensen
- Molecular Pharmacology Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Amanda Krøger Johansen
- Molecular Pharmacology Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Mikela Reinholdt Zinn
- Molecular Pharmacology Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Hulda Maria Jensdóttir
- Molecular Pharmacology Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - John Dirk Vestergaard Nieland
- 2N Pharma ApS, NOVI Science Park, Aalborg, Denmark
- Molecular Pharmacology Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
7
|
Battaglini M, Marino A, Montorsi M, Carmignani A, Ceccarelli MC, Ciofani G. Nanomaterials as Microglia Modulators in the Treatment of Central Nervous System Disorders. Adv Healthc Mater 2024; 13:e2304180. [PMID: 38112345 DOI: 10.1002/adhm.202304180] [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: 12/11/2023] [Indexed: 12/21/2023]
Abstract
Microglia play a pivotal role in the central nervous system (CNS) homeostasis, acting as housekeepers and defenders of the surrounding environment. These cells can elicit their functions by shifting into two main phenotypes: pro-inflammatory classical phenotype, M1, and anti-inflammatory alternative phenotype, M2. Despite their pivotal role in CNS homeostasis, microglia phenotypes can influence the development and progression of several CNS disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, ischemic stroke, traumatic brain injuries, and even brain cancer. It is thus clear that the possibility of modulating microglia activation has gained attention as a therapeutic tool against many CNS pathologies. Nanomaterials are an unprecedented tool for manipulating microglia responses, in particular, to specifically target microglia and elicit an in situ immunomodulation activity. This review focuses the discussion on two main aspects: analyzing the possibility of using nanomaterials to stimulate a pro-inflammatory response of microglia against brain cancer and introducing nanostructures able to foster an anti-inflammatory response for treating neurodegenerative disorders. The final aim is to stimulate the analysis of the development of new microglia nano-immunomodulators, paving the way for innovative and effective therapeutic approaches for the treatment of CNS disorders.
Collapse
Affiliation(s)
- Matteo Battaglini
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Margherita Montorsi
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Alessio Carmignani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Maria Cristina Ceccarelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
- Scuola Superiore Sant'Anna, The BioRobotics Institute, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera, 56025, Italy
| |
Collapse
|
8
|
Oizumi H, Miyamoto Y, Seiwa C, Yamamoto M, Yoshioka N, Iizuka S, Torii T, Ohbuchi K, Mizoguchi K, Yamauchi J, Asou H. Lethal adulthood myelin breakdown by oligodendrocyte-specific Ddx54 knockout. iScience 2023; 26:107448. [PMID: 37720086 PMCID: PMC10502337 DOI: 10.1016/j.isci.2023.107448] [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: 02/21/2022] [Revised: 05/08/2023] [Accepted: 07/18/2023] [Indexed: 09/19/2023] Open
Abstract
Multiple sclerosis (MS) is a leading disease that causes disability in young adults. We have previously shown that a DEAD-box RNA helicase Ddx54 binds to mRNA and protein isoforms of myelin basic protein (MBP) and that Ddx54 siRNA blocking abrogates oligodendrocyte migration and myelination. Herein, we show that MBP-driven Ddx54 knockout mice (Ddx54 fl/fl;MBP-Cre), after the completion of normal postnatal myelination, gradually develop abnormalities in behavioral profiles and learning ability, inner myelin sheath breakdown, loss of myelinated axons, apoptosis of oligodendrocytes, astrocyte and microglia activation, and they die within 7 months but show minimal peripheral immune cell infiltration. Myelin in Ddx54fl/fl;MBP-Cre is highly vulnerable to the neurotoxicant cuprizone and Ddx54 knockdown greatly impairs myelination in vitro. Ddx54 expression in oligodendrocyte-lineage cells decreased in corpus callosum of MS patients. Our results demonstrate that Ddx54 is indispensable for myelin homeostasis, and they provide a demyelinating disease model based on intrinsic disintegration of adult myelin.
Collapse
Affiliation(s)
- Hiroaki Oizumi
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | - Yuki Miyamoto
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Chika Seiwa
- Glovia Myelin Research Institute, Tsurumi-ku, Yokohama, Kanagawa 230-0046, Japan
| | - Masahiro Yamamoto
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | - Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Seiichi Iizuka
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | - Tomohiro Torii
- Laboratory of Ion Channel Pathophysiology, Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Katsuya Ohbuchi
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | | | - Junji Yamauchi
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Hiroaki Asou
- Glovia Myelin Research Institute, Tsurumi-ku, Yokohama, Kanagawa 230-0046, Japan
| |
Collapse
|
9
|
Fodder K, de Silva R, Warner TT, Bettencourt C. The contribution of DNA methylation to the (dys)function of oligodendroglia in neurodegeneration. Acta Neuropathol Commun 2023; 11:106. [PMID: 37386505 PMCID: PMC10311741 DOI: 10.1186/s40478-023-01607-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023] Open
Abstract
Neurodegenerative diseases encompass a heterogeneous group of conditions characterised by the progressive degeneration of the structure and function of the central or peripheral nervous systems. The pathogenic mechanisms underlying these diseases are not fully understood. However, a central feature consists of regional aggregation of proteins in the brain, such as the accumulation of β-amyloid plaques in Alzheimer's disease (AD), inclusions of hyperphosphorylated microtubule-binding tau in AD and other tauopathies, or inclusions containing α-synuclein in Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). Various pathogenic mechanisms are thought to contribute to disease, and an increasing number of studies implicate dysfunction of oligodendrocytes (the myelin producing cells of the central nervous system) and myelin loss. Aberrant DNA methylation, the most widely studied epigenetic modification, has been associated with many neurodegenerative diseases, including AD, PD, DLB and MSA, and recent findings highlight aberrant DNA methylation in oligodendrocyte/myelin-related genes. Here we briefly review the evidence showing that changes to oligodendrocytes and myelin are key in neurodegeneration, and explore the relevance of DNA methylation in oligodendrocyte (dys)function. As DNA methylation is reversible, elucidating its involvement in pathogenic mechanisms of neurodegenerative diseases and in dysfunction of specific cell-types such as oligodendrocytes may bring opportunities for therapeutic interventions for these diseases.
Collapse
Affiliation(s)
- Katherine Fodder
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Rohan de Silva
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Thomas T Warner
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Conceição Bettencourt
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
10
|
Dolma S, Joshi A. The Node of Ranvier as an Interface for Axo-Glial Interactions: Perturbation of Axo-Glial Interactions in Various Neurological Disorders. J Neuroimmune Pharmacol 2023; 18:215-234. [PMID: 37285016 DOI: 10.1007/s11481-023-10072-z] [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: 09/08/2022] [Accepted: 05/19/2023] [Indexed: 06/08/2023]
Abstract
The action potential conduction along the axon is highly dependent on the healthy interactions between the axon and myelin-producing glial cells. Myelin, which facilitates action potential, is the protective insulation around the axon formed by Schwann cells and oligodendrocytes in the peripheral (PNS) and central nervous system (CNS), respectively. Myelin is a continuous structure with intermittent gaps called nodes of Ranvier, which are the sites enriched with ion channels, transmembrane, scaffolding, and cytoskeletal proteins. Decades-long extensive research has identified a comprehensive proteome with strictly regularized localization at the node of Ranvier. Concurrently, axon-glia interactions at the node of Ranvier have gathered significant attention as the pathophysiological targets for various neurodegenerative disorders. Numerous studies have shown the alterations in the axon-glia interactions culminating in neurological diseases. In this review, we have provided an update on the molecular composition of the node of Ranvier. Further, we have discussed in detail the consequences of disruption of axon-glia interactions during the pathogenesis of various CNS and PNS disorders.
Collapse
Affiliation(s)
- Sonam Dolma
- Department of Pharmacy, Birla Institute of Technology and Sciences- Pilani, Hyderabad campus, Telangana state, India
| | - Abhijeet Joshi
- Department of Pharmacy, Birla Institute of Technology and Sciences- Pilani, Hyderabad campus, Telangana state, India.
| |
Collapse
|
11
|
Neel DV, Basu H, Gunner G, Chiu IM. Catching a killer: Mechanisms of programmed cell death and immune activation in Amyotrophic Lateral Sclerosis. Immunol Rev 2022; 311:130-150. [PMID: 35524757 PMCID: PMC9489610 DOI: 10.1111/imr.13083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 04/21/2022] [Indexed: 12/13/2022]
Abstract
In the central nervous system (CNS), execution of programmed cell death (PCD) is crucial for proper neurodevelopment. However, aberrant activation of these pathways in adult CNS leads to neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). How a cell dies is critical, as it can drive local immune activation and tissue damage. Classical apoptosis engages several mechanisms to evoke "immunologically silent" responses, whereas other forms of programmed death such as pyroptosis, necroptosis, and ferroptosis release molecules that can potentiate immune responses and inflammation. In ALS, a fatal neuromuscular disorder marked by progressive death of lower and upper motor neurons, several cell types in the CNS express machinery for multiple PCD pathways. The specific cell types engaging PCD, and ultimate mechanisms by which neuronal death occurs in ALS are not well defined. Here, we provide an overview of different PCD pathways implicated in ALS. We also examine immune activation in ALS and differentiate apoptosis from necrotic mechanisms based on downstream immunological consequences. Lastly, we highlight therapeutic strategies that target cell death pathways in the treatment of neurodegeneration and inflammation in ALS.
Collapse
Affiliation(s)
- Dylan V Neel
- Harvard Medical School, Department of Immunology, Blavatnik Institute, Boston, MA, USA
| | - Himanish Basu
- Harvard Medical School, Department of Immunology, Blavatnik Institute, Boston, MA, USA
| | - Georgia Gunner
- Harvard Medical School, Department of Immunology, Blavatnik Institute, Boston, MA, USA
| | - Isaac M Chiu
- Harvard Medical School, Department of Immunology, Blavatnik Institute, Boston, MA, USA
- Lead contact
| |
Collapse
|
12
|
Yusuf IO, Qiao T, Parsi S, Tilvawala R, Thompson PR, Xu Z. Protein citrullination marks myelin protein aggregation and disease progression in mouse ALS models. Acta Neuropathol Commun 2022; 10:135. [PMID: 36076282 PMCID: PMC9458309 DOI: 10.1186/s40478-022-01433-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
Increased protein citrullination (PC) and dysregulated protein arginine deiminase (PAD) activity have been observed in several neurodegenerative diseases. PC is a posttranslational modification catalyzed by the PADs. PC converts peptidyl-arginine to peptidyl-citrulline, thereby reducing the positive charges and altering structure and function of proteins. Of the five PADs, PAD2 is the dominant isoform in the central nervous system (CNS). Abnormal PC and PAD dysregulation are associated with numerous pathological conditions, including inflammatory diseases and neurodegeneration. Animal model studies have shown therapeutic efficacy from inhibition of PADs, thus suggesting a role of PC in pathogenesis. To determine whether PC contribute to amyotrophic lateral sclerosis (ALS), a deadly neurodegenerative disease characterized by loss of motor neurons, paralysis, and eventual death, we investigated alterations of PC and PAD2 in two different transgenic mouse models of ALS expressing human mutant SOD1G93A and PFN1C71G, respectively. PC and PAD2 expression are altered dynamically in the spinal cord during disease progression in both models. PC and PAD2 increase progressively in astrocytes with the development of reactive astrogliosis, while decreasing in neurons. Importantly, in the spinal cord white matter, PC accumulates in protein aggregates that contain the myelin proteins PLP and MBP. PC also accumulates progressively in insoluble protein fractions during disease progression. Finally, increased PC and PAD2 expression spatially correlate with areas of the CNS with the most severe motor neuron degeneration. These results suggest that altered PC is an integral part of the neurodegenerative process and potential biomarkers for disease progression in ALS. Moreover, increased PC may contribute to disease-associated processes such as myelin protein aggregation, myelin degeneration, and astrogliosis.
Collapse
Affiliation(s)
- Issa O. Yusuf
- grid.168645.80000 0001 0742 0364Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, MA 01605 USA
| | - Tao Qiao
- grid.168645.80000 0001 0742 0364Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, MA 01605 USA ,grid.423286.90000 0004 0507 1326Present Address: Astellas Pharma, 33 Locke Dr, Marlborough, MA 01752 USA
| | - Sepideh Parsi
- grid.168645.80000 0001 0742 0364Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, MA 01605 USA ,grid.38142.3c000000041936754XPresent Address: Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| | - Ronak Tilvawala
- grid.168645.80000 0001 0742 0364Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, MA 01605 USA ,grid.509226.aPresent Address: Scorpion Therapeutics, 1 Winthrop Square, Boston, MA 02110 USA
| | - Paul R. Thompson
- grid.168645.80000 0001 0742 0364Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, MA 01605 USA ,grid.168645.80000 0001 0742 0364Program in Chemical Biology, University of Massachusetts Medical School, Worcester, MA 01605 USA
| | - Zuoshang Xu
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
13
|
Ozzoude M, Varriano B, Beaton D, Ramirez J, Holmes MF, Scott CJM, Gao F, Sunderland KM, McLaughlin P, Rabin J, Goubran M, Kwan D, Roberts A, Bartha R, Symons S, Tan B, Swartz RH, Abrahao A, Saposnik G, Masellis M, Lang AE, Marras C, Zinman L, Shoesmith C, Borrie M, Fischer CE, Frank A, Freedman M, Montero-Odasso M, Kumar S, Pasternak S, Strother SC, Pollock BG, Rajji TK, Seitz D, Tang-Wai DF, Turnbull J, Dowlatshahi D, Hassan A, Casaubon L, Mandzia J, Sahlas D, Breen DP, Grimes D, Jog M, Steeves TDL, Arnott SR, Black SE, Finger E, Tartaglia MC. Investigating the contribution of white matter hyperintensities and cortical thickness to empathy in neurodegenerative and cerebrovascular diseases. GeroScience 2022; 44:1575-1598. [PMID: 35294697 DOI: 10.1007/s11357-022-00539-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
Change in empathy is an increasingly recognised symptom of neurodegenerative diseases and contributes to caregiver burden and patient distress. Empathy impairment has been associated with brain atrophy but its relationship to white matter hyperintensities (WMH) is unknown. We aimed to investigate the relationships amongst WMH, brain atrophy, and empathy deficits in neurodegenerative and cerebrovascular diseases. Five hundred thirteen participants with Alzheimer's disease/mild cognitive impairment, amyotrophic lateral sclerosis, frontotemporal dementia (FTD), Parkinson's disease, or cerebrovascular disease (CVD) were included. Empathy was assessed using the Interpersonal Reactivity Index. WMH were measured using a semi-automatic segmentation and FreeSurfer was used to measure cortical thickness. A heterogeneous pattern of cortical thinning was found between groups, with FTD showing thinning in frontotemporal regions and CVD in left superior parietal, left insula, and left postcentral. Results from both univariate and multivariate analyses revealed that several variables were associated with empathy, particularly cortical thickness in the fronto-insulo-temporal and cingulate regions, sex (female), global cognition, and right parietal and occipital WMH. Our results suggest that cortical atrophy and WMH may be associated with empathy deficits in neurodegenerative and cerebrovascular diseases. Future work should consider investigating the longitudinal effects of WMH and atrophy on empathy deficits in neurodegenerative and cerebrovascular diseases.
Collapse
Affiliation(s)
- Miracle Ozzoude
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th floor 6KD-407, Toronto, ON, M5T 0S8, Canada.,L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Brenda Varriano
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th floor 6KD-407, Toronto, ON, M5T 0S8, Canada
| | - Derek Beaton
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada
| | - Joel Ramirez
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Melissa F Holmes
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Christopher J M Scott
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Fuqiang Gao
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | - Paula McLaughlin
- Nova Scotia Health and Dalhousie University, Halifax, NS, Canada
| | - Jennifer Rabin
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Maged Goubran
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Donna Kwan
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.,Queen's University, Kingston, ON, Canada
| | - Angela Roberts
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA.,School of Communication Sciences and Disorders, Faculty of Health Sciences, Western University, London, ON, Canada
| | - Robert Bartha
- Robarts Research Institute, Western University, London, ON, Canada
| | - Sean Symons
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Brian Tan
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada
| | - Richard H Swartz
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Agessandro Abrahao
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Gustavo Saposnik
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Mario Masellis
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Anthony E Lang
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Edmond J Safra Program for Parkinson Disease, Movement Disorder Clinic, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Connie Marras
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Edmond J Safra Program for Parkinson Disease, Movement Disorder Clinic, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Lorne Zinman
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Christen Shoesmith
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada
| | - Michael Borrie
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,St. Joseph's Healthcare Centre, London, ON, Canada
| | - Corinne E Fischer
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Andrew Frank
- Department of Medicine (Neurology), University of Ottawa Brain and Mind Research Institute and Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Bruyère Research Institute, Ottawa, ON, Canada
| | - Morris Freedman
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Neurology, Baycrest Health Sciences, Toronto, ON, Canada
| | - Manuel Montero-Odasso
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Lawson Health Research Institute, London, ON, Canada.,Gait and Brain Lab, Parkwood Institute, London, ON, Canada
| | - Sanjeev Kumar
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Adult Neurodevelopment and Geriatric Psychiatry, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Stephen Pasternak
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada
| | - Stephen C Strother
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Bruce G Pollock
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Adult Neurodevelopment and Geriatric Psychiatry, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tarek K Rajji
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Adult Neurodevelopment and Geriatric Psychiatry, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada
| | - Dallas Seitz
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - David F Tang-Wai
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Memory Clinic, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - John Turnbull
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Dar Dowlatshahi
- Department of Medicine (Neurology), University of Ottawa Brain and Mind Research Institute and Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Ayman Hassan
- Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada
| | - Leanne Casaubon
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jennifer Mandzia
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada.,Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Demetrios Sahlas
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - David P Breen
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK.,Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - David Grimes
- Department of Medicine (Neurology), University of Ottawa Brain and Mind Research Institute and Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Mandar Jog
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada.,Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,London Health Sciences Centre, London, ON, Canada
| | - Thomas D L Steeves
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Stephen R Arnott
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada
| | - Sandra E Black
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada.,Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | | | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th floor 6KD-407, Toronto, ON, M5T 0S8, Canada. .,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada. .,Memory Clinic, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
14
|
Liu YF, Zou ZY, Cai LM, Lin JH, Zhou MX, Huang NX, Zhan C, Chen HJ. Characterizing Sensorimotor-Related Area Abnormalities in Amyotrophic Lateral Sclerosis: An Intravoxel Incoherent Motion Magnetic Resonance Imaging Study. Acad Radiol 2022; 29 Suppl 3:S141-S146. [PMID: 34481706 DOI: 10.1016/j.acra.2021.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/08/2021] [Accepted: 07/18/2021] [Indexed: 12/12/2022]
Abstract
RATIONALE AND OBJECTIVES To investigate the microperfusion and water molecule diffusion alterations in sensorimotor-related areas in amyotrophic lateral sclerosis (ALS) using intravoxel incoherent motion (IVIM) magnetic resonance imaging. MATERIALS AND METHODS IVIM data were obtained from 43 ALS patients and 31 controls. This study employed the revised ALS Functional Rating Scale (ALSFRS-R) in evaluating disease severity. IVIM-derived metrics were calculated, including diffusion coefficient (D), pseudo-diffusion coefficient, and perfusion fraction. Conventional apparent diffusion coefficient was also computed. Atlas-based analysis was conducted to detect between-group difference in these metrics in sensorimotor-related gray/white matter areas. Spearman correlation analysis was employed to establish correlation between various metrics and ALSFRS-R. RESULTS ALS patients had perfusion fraction (× 10-3) reduction in the left presupplementary motor area (60.72 ± 16.15 vs. 71.15 ± 12.98, p = 0.016), right presupplementary motor area (61.35 ± 17.02 vs. 72.18 ± 14.22, p = 0.016), left supplementary motor area (55.73 ± 12.29 vs. 64.12 ± 9.17, p = 0.015), and right supplementary motor area (56.53 ± 11.93 vs. 63.67 ± 10.03, p = 0.020). Patients showed D (× 10-6 mm2/s) increase in a white matter tract projecting to the right ventral premotor cortex (714.20 ± 39.75 vs. 691.01 ± 24.53, p = 0.034). A negative correlation between D of right ventral premotor cortex tract and ALSFRS-R score was observed (r = -0.316, p = 0.039). CONCLUSION These findings suggest aberrant microperfusion and water molecule diffusion in the sensorimotor-related areas in ALS patients, which are associated with motor impairment in ALS.
Collapse
Affiliation(s)
- Yuan-Fen Liu
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhang-Yu Zou
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Li-Min Cai
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Jia-Hui Lin
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Min-Xiong Zhou
- College of Medical Imaging, Shang Hai University of Medicine & Health Sciences, Shanghai, China
| | - Nao-Xin Huang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Chuanyin Zhan
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Hua-Jun Chen
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| |
Collapse
|
15
|
Siokas V, Aloizou A, Liampas I, Bakirtzis C, Tsouris Z, Sgantzos M, Liakos P, Bogdanos DP, Hadjigeorgiou GM, Dardiotis E. Myelin-associated oligodendrocyte basic protein rs616147 polymorphism as a risk factor for Parkinson's disease. Acta Neurol Scand 2022; 145:223-228. [PMID: 34694630 DOI: 10.1111/ane.13538] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/25/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The rs616147 polymorphism of the myelin-associated oligodendrocyte basic protein (MOBP) gene locus has been associated with amyotrophic lateral sclerosis (ALS). ALS and Parkinson's disease (PD) are two common neurodegenerative disorders that share features regarding their etiology, pathophysiology, and genetic backgrounds. While the MOBP rs616147 polymorphism has been associated with ALS, little is known about its role in PD. OBJECTIVE To assess the role of MOBP rs616147 on PD risk. METHODS This case-control comparison study consists of 358 PD-affected cases and 358 controls from the Neurology Clinic of the University Hospital of Larissa, University of Thessaly, Faculty of Medicine, in Greece. The diagnosis of PD was made by a specialist neurologist according to the UK Parkinson's Disease Society Brain Bank's clinical criteria. All the participants were genotyped for the MOBP rs616147. Furthermore, in order to validate our results, we genotyped 327 patients with Alzheimer's disease (AD) for MOBP rs616147 and compared them with the control group. RESULTS According to the univariate analysis, there was a significant association between rs616147 and PD in the dominant (OR [95% C.I.] = 0.70 [0.52-0.94], p = .018), the overdominant (OR [95% C.I.] = 0.68 [0.50-0.92], p = .011), and in the codominant (G/A VS G/G; OR [95% C.I.] = 0.66 [0.48-0.91], p = .035) modes of inheritance. In contrast, there was no association between the MOBP rs616147 polymorphism and AD. CONCLUSIONS We provide preliminary results associating MOBP rs616147 genetic variant with PD.
Collapse
Affiliation(s)
- Vasileios Siokas
- Laboratory of Neurogenetics Department of Neurology University Hospital of Larissa Faculty of Medicine School of Health Sciences Larissa Greece
| | - Athina‐Maria Aloizou
- Laboratory of Neurogenetics Department of Neurology University Hospital of Larissa Faculty of Medicine School of Health Sciences Larissa Greece
| | - Ioannis Liampas
- Laboratory of Neurogenetics Department of Neurology University Hospital of Larissa Faculty of Medicine School of Health Sciences Larissa Greece
| | - Christos Bakirtzis
- B' Department of Neurology Multiple Sclerosis Center AHEPA University Hospital Aristotle University of Thessaloniki Thessaloniki Greece
| | - Zisis Tsouris
- Laboratory of Neurogenetics Department of Neurology University Hospital of Larissa Faculty of Medicine School of Health Sciences Larissa Greece
| | - Markos Sgantzos
- Laboratory of Neurogenetics Department of Neurology University Hospital of Larissa Faculty of Medicine School of Health Sciences Larissa Greece
| | - Panagiotis Liakos
- Laboratory of Biochemistry Faculty of Medicine University of Thessaly Larissa Greece
| | - Dimitrios P. Bogdanos
- Faculty of Medicine Department of Rheumatology and Clinical Immunology University General Hospital of Larissa School of Health Sciences University of Thessaly Larissa Greece
| | - Georgios M. Hadjigeorgiou
- Laboratory of Neurogenetics Department of Neurology University Hospital of Larissa Faculty of Medicine School of Health Sciences Larissa Greece
- Department of Neurology Medical School University of Cyprus Nicosia Cyprus
| | - Efthimios Dardiotis
- Laboratory of Neurogenetics Department of Neurology University Hospital of Larissa Faculty of Medicine School of Health Sciences Larissa Greece
| |
Collapse
|
16
|
White matter microglia heterogeneity in the CNS. Acta Neuropathol 2022; 143:125-141. [PMID: 34878590 DOI: 10.1007/s00401-021-02389-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/17/2021] [Accepted: 11/28/2021] [Indexed: 02/07/2023]
Abstract
Microglia, the resident myeloid cells in the central nervous system (CNS) play critical roles in shaping the brain during development, responding to invading pathogens, and clearing tissue debris or aberrant protein aggregations during ageing and neurodegeneration. The original concept that like macrophages, microglia are either damaging (pro-inflammatory) or regenerative (anti-inflammatory) has been updated to a kaleidoscope view of microglia phenotypes reflecting their wide-ranging roles in maintaining homeostasis in the CNS and, their contribution to CNS diseases, as well as aiding repair. The use of new technologies including single cell/nucleus RNA sequencing has led to the identification of many novel microglia states, allowing for a better understanding of their complexity and distinguishing regional variations in the CNS. This has also revealed differences between species and diseases, and between microglia and other myeloid cells in the CNS. However, most of the data on microglia heterogeneity have been generated on cells isolated from the cortex or whole brain, whereas white matter changes and differences between white and grey matter have been relatively understudied. Considering the importance of microglia in regulating white matter health, we provide a brief update on the current knowledge of microglia heterogeneity in the white matter, how microglia are important for the development of the CNS, and how microglial ageing affects CNS white matter homeostasis. We discuss how microglia are intricately linked to the classical white matter diseases such as multiple sclerosis and genetic white matter diseases, and their putative roles in neurodegenerative diseases in which white matter is also affected. Understanding the wide variety of microglial functions in the white matter may provide the basis for microglial targeted therapies for CNS diseases.
Collapse
|
17
|
Martinsen V, Kursula P. Multiple sclerosis and myelin basic protein: insights into protein disorder and disease. Amino Acids 2021; 54:99-109. [PMID: 34889995 PMCID: PMC8810476 DOI: 10.1007/s00726-021-03111-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/24/2021] [Indexed: 01/18/2023]
Abstract
Myelin basic protein (MBP) is an abundant protein in central nervous system (CNS) myelin. MBP has long been studied as a factor in the pathogenesis of the autoimmune neurodegenerative disease multiple sclerosis (MS). MS is characterized by CNS inflammation, demyelination, and axonal loss. One of the main theories on the pathogenesis of MS suggests that exposure to foreign antigens causes the activation of cross-reactive T cells in genetically susceptible individuals, with MBP being a possible autoantigen. While a direct role for MBP as a primary antigen in human MS is unclear, it is clear that MBP and its functions in myelin formation and long-term maintenance are linked to MS. This review looks at some key molecular characteristics of MBP and its relevance to MS, as well as the mechanisms of possible molecular mimicry between MBP and some viral antigens. We also discuss the use of serum anti-myelin antibodies as biomarkers for disease. MBP is a prime example of an apparently simple, but in fact biochemically and structurally complex molecule, which is closely linked to both normal nervous system development and neurodegenerative disease.
Collapse
Affiliation(s)
- Vebjørn Martinsen
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5020, Bergen, Norway
| | - Petri Kursula
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5020, Bergen, Norway. .,Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220, Oulu, Finland.
| |
Collapse
|
18
|
MOBP rs616147 Polymorphism and Risk of Amyotrophic Lateral Sclerosis in a Greek Population: A Case-Control Study. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57121337. [PMID: 34946282 PMCID: PMC8708438 DOI: 10.3390/medicina57121337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 02/05/2023]
Abstract
Background and Objectives: To date, only one study has investigated the association between the rs616147 polymorphism of the Myelin-associated Oligodendrocyte Basic Protein (MOBP) locus and Amyotrophic Lateral Sclerosis (ALS). Materials and Methods: A case-control study was performed. Patients with definite sporadic ALS were prospectively and consecutively recruited from the inpatient and outpatient clinics of the Neurology Department of the General University Hospital of Larissa, Central Greece. Community based, age and sex matched healthy individuals with a free personal and family history constituted the control group. Results: A total of 155 patients with definite sporadic ALS and an equal number of healthy controls were genotyped. The power of our sample size was slightly above 80% and MOBP rs616147 was determined to be in Hardy-Weinberg Equilibrium among healthy participants (p = 1.00). According to the univariate analysis, there was no significant relationship between rs616147 and ALS [log-additive OR = 0.85 (0.61, 1.19), over-dominant OR = 0.73 (0.46, 1.15), recessive OR = 1.02 (0.50, 2.09), dominant OR = 0.74 (0.47, 1.16), co-dominant OR1 = 0.71 (0.44, 1.14) and co-dominant OR2 = 0.88 (0.42, 1.84). Additionally, the effect of rs616147 on the age of ALS onset was determined insignificant using both unadjusted and adjusted (sex, site of onset) cox-proportional models. Finally, rs616147 was not related to the site of ALS onset. Conclusions: Our study is the first to report the absence of an association between MOBP rs616147 and ALS among individuals of Greek ancestry. Additional, larger nationwide and multi-ethnic studies are warranted to shed light on the connection between rs616147 and ALS.
Collapse
|
19
|
Bono S, Feligioni M, Corbo M. Impaired antioxidant KEAP1-NRF2 system in amyotrophic lateral sclerosis: NRF2 activation as a potential therapeutic strategy. Mol Neurodegener 2021; 16:71. [PMID: 34663413 PMCID: PMC8521937 DOI: 10.1186/s13024-021-00479-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Oxidative stress (OS) is an imbalance between oxidant and antioxidant species and, together with other numerous pathological mechanisms, leads to the degeneration and death of motor neurons (MNs) in amyotrophic lateral sclerosis (ALS). MAIN BODY Two of the main players in the molecular and cellular response to OS are NRF2, the transcription nuclear factor erythroid 2-related factor 2, and its principal negative regulator, KEAP1, Kelch-like ECH (erythroid cell-derived protein with CNC homology)-associated protein 1. Here we first provide an overview of the structural organization, regulation, and critical role of the KEAP1-NRF2 system in counteracting OS, with a focus on its alteration in ALS. We then examine several compounds capable of promoting NRF2 activity thereby inducing cytoprotective effects, and which are currently in different stages of clinical development for many pathologies, including neurodegenerative diseases. CONCLUSIONS Although challenges associated with some of these compounds remain, important advances have been made in the development of safer and more effective drugs that could actually represent a breakthrough for fatal degenerative diseases such as ALS.
Collapse
Affiliation(s)
- Silvia Bono
- Need Institute, Laboratory of Neurobiology for Translational Medicine, c/o Casa di Cura del Policlinico (CCP), Via Dezza 48, 20144 Milan, Italy
| | - Marco Feligioni
- Need Institute, Laboratory of Neurobiology for Translational Medicine, c/o Casa di Cura del Policlinico (CCP), Via Dezza 48, 20144 Milan, Italy
- Laboratory of Neuronal Cell Signaling, EBRI Rita Levi-Montalcini Foundation, 00161 Rome, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa di Cura del Policlinico (CCP), Via Dezza 48, 20144 Milan, Italy
| |
Collapse
|
20
|
Ogura A, Kawabata K, Watanabe H, Choy SW, Bagarinao E, Kato T, Imai K, Masuda M, Ohdake R, Hara K, Nakamura R, Atsuta N, Nakamura T, Katsuno M, Sobue G. Fiber-specific white matter analysis reflects upper motor neuron impairment in amyotrophic lateral sclerosis. Eur J Neurol 2021; 29:432-440. [PMID: 34632672 DOI: 10.1111/ene.15136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/30/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE To clarify the relationship between fiber-specific white matter changes in amyotrophic lateral sclerosis (ALS) and clinical signs of upper motor neuron (UMN) involvement, we performed a fixel-based analysis (FBA), a novel framework for diffusion-weighted imaging analysis. METHODS We enrolled 96 participants, including 48 nonfamilial ALS patients and 48 age- and sex-matched healthy controls (HCs), in this study and conducted whole-brain FBA and voxel-based morphometry analysis. We compared the fiber density (FD), fiber morphology (fiber cross-section [FC]), and a combined index of FD and FC (FDC) between the ALS and HC groups. We performed a tract-of-interest analysis to extract FD values across the significant regions in the whole-brain analysis. Then, we evaluated the associations between FD values and clinical variables. RESULTS The bilateral corticospinal tracts (CSTs) and the corpus callosum (CC) showed reduced FD and FDC in ALS patients compared with HCs (p < 0.05, familywise error-corrected), and the comparison of FCs revealed no region that was significantly different from another. Voxel-based morphometry showed cortical volume reduction in the regions, including the primary motor area. Clinical scores showed correlations with FD values in the CSTs (UMN score: rho = -0.530, p < 0.001; central motor conduction time [CMCT] in the upper limb: rho = -0.474, p = 0.008; disease duration: rho = -0.383, p = 0.007; ALS Functional Rating Scale-Revised: rho = 0.340, p = 0.018). In addition, patients whose CMCT was not calculated due to unevoked waves also showed FD reduction in the CSTs. CONCLUSIONS Our findings suggest that FD values in the CST estimated via FBA can be potentially used in evaluating UMN impairments.
Collapse
Affiliation(s)
- Aya Ogura
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuya Kawabata
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hirohisa Watanabe
- Brain and Mind Research Center, Nagoya University, Nagoya, Japan.,Department of Neurology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Shao Wei Choy
- Center for Intelligent Signal and Imaging Research, Universiti Teknologi PETRONAS, Seri Iskandar, Malaysia
| | - Epifanio Bagarinao
- Brain and Mind Research Center, Nagoya University, Nagoya, Japan.,Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshiyasu Kato
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazunori Imai
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michihito Masuda
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Reiko Ohdake
- Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| | - Kazuhiro Hara
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryoichi Nakamura
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoki Atsuta
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomohiko Nakamura
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Gen Sobue
- Brain and Mind Research Center, Nagoya University, Nagoya, Japan.,Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Aichi Medical University, Nagakute, Japan
| |
Collapse
|
21
|
Balestri S, Del Giovane A, Sposato C, Ferrarelli M, Ragnini-Wilson A. The Current Challenges for Drug Discovery in CNS Remyelination. Int J Mol Sci 2021; 22:ijms22062891. [PMID: 33809224 PMCID: PMC8001072 DOI: 10.3390/ijms22062891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
The myelin sheath wraps around axons, allowing saltatory currents to be transmitted along neurons. Several genetic, viral, or environmental factors can damage the central nervous system (CNS) myelin sheath during life. Unless the myelin sheath is repaired, these insults will lead to neurodegeneration. Remyelination occurs spontaneously upon myelin injury in healthy individuals but can fail in several demyelination pathologies or as a consequence of aging. Thus, pharmacological intervention that promotes CNS remyelination could have a major impact on patient’s lives by delaying or even preventing neurodegeneration. Drugs promoting CNS remyelination in animal models have been identified recently, mostly as a result of repurposing phenotypical screening campaigns that used novel oligodendrocyte cellular models. Although none of these have as yet arrived in the clinic, promising candidates are on the way. Many questions remain. Among the most relevant is the question if there is a time window when remyelination drugs should be administrated and why adult remyelination fails in many neurodegenerative pathologies. Moreover, a significant challenge in the field is how to reconstitute the oligodendrocyte/axon interaction environment representative of healthy as well as disease microenvironments in drug screening campaigns, so that drugs can be screened in the most appropriate disease-relevant conditions. Here we will provide an overview of how the field of in vitro models developed over recent years and recent biological findings about how oligodendrocytes mature after reactivation of their staminal niche. These data have posed novel questions and opened new views about how the adult brain is repaired after myelin injury and we will discuss how these new findings might change future drug screening campaigns for CNS regenerative drugs.
Collapse
|
22
|
Basso MR, Whiteside D, Combs D, Woods SP, Hoffmeister J, Mulligan R, Arnett P, Alden E, Tobin O. Memory in multiple sclerosis: A reappraisal using the item specific deficit approach. Neuropsychology 2021; 35:207-219. [PMID: 33764111 PMCID: PMC8396077 DOI: 10.1037/neu0000712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE As many as 65% of people with multiple sclerosis (MS) have clinically significant memory impairment, but the nature of this deficit is controversial. Some investigations suggest that an inability to retrieve newly learned information from memory is prominent, whereas others imply that compromised acquisition accounts for impairment. Prior research has not simultaneously evaluated acquisition and retrieval processes in MS, and fewer have attempted to account for initial acquisition when studying retrieval. The Item Specific Deficit Approach (ISDA) offers a method of quantifying acquisition, retrieval, and retention processes, with the latter two mechanisms being adjusted for initial acquisition. To simultaneously quantify acquisition and retrieval abilities, the ISDA was applied to list learning performance in two independent samples of people with MS and corresponding healthy comparison groups. PARTICIPANTS AND METHODS Study 1 included 85 people with MS and 47 healthy individuals. Study 2 involved a separate sample of 79 people with MS and 22 healthy people. They were administered neuropsychological batteries, and participants with MS were classified as globally impaired or unimpaired. The California Verbal Learning Test-II was administered to assess new-learning in both studies, and responses were scored using the ISDA. RESULTS Both studies revealed that cognitively impaired people with MS manifest weaknesses involving acquisition and retrieval. Nearly identical effect sizes emerged across samples, with cognitive impairment achieving a medium effect upon acquisition and a large effect upon retrieval. CONCLUSIONS These findings accord well with previous research showing diminished acquisition and retrieval among people with MS. The results may also reconcile contradictory findings in the extant literature by showing that memory impairment in MS is not exclusively attributable to either acquisition or retrieval. Rather, both processes may manifest across people with MS. The replication across samples with nearly identical effect sizes implies that these effects are reliable and possess external validity. These data hold implications for memory rehabilitation interventions involving people with MS, and suggest that acquisition and retrieval processes should be addressed in treatment. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
|
23
|
AAV9-mediated gene delivery of MCT1 to oligodendrocytes does not provide a therapeutic benefit in a mouse model of ALS. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:508-519. [PMID: 33614825 PMCID: PMC7878966 DOI: 10.1016/j.omtm.2021.01.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/09/2021] [Indexed: 12/11/2022]
Abstract
Oligodendrocyte dysfunction has been implicated in the pathophysiology of amyotrophic lateral sclerosis (ALS), a neurodegenerative disorder characterized by progressive motor neuron loss. The failure of trophic support provided by oligodendrocytes is associated with a concomitant reduction in oligodendroglial monocarboxylate transporter 1 (MCT1) expression and is detrimental for the long-term survival of motor neuron axons. Therefore, we established an adeno-associated virus 9 (AAV9)-based platform by which MCT1 was targeted mostly to white matter oligodendrocytes to investigate whether this approach could provide a therapeutic benefit in the SOD1G93A mouse model of ALS. Despite good oligodendrocyte transduction and AAV-mediated MCT1 transgene expression, the disease outcome of SOD1G93A mice was not altered. Our study further increases our current understanding about the complex nature of oligodendrocyte pathology in ALS and provides valuable insights into the future development of therapeutic strategies to efficiently modulate these cells.
Collapse
|
24
|
Pro-Inflammatory Signaling Upregulates a Neurotoxic Conotoxin-Like Protein Encrypted Within Human Endogenous Retrovirus-K. Cells 2020; 9:cells9071584. [PMID: 32629888 PMCID: PMC7407490 DOI: 10.3390/cells9071584] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/20/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
Motor neuron degeneration and spinal cord demyelination are hallmark pathological events in Amyotrophic Lateral Sclerosis (ALS). Endogenous retrovirus-K (ERVK) expression has an established association with ALS neuropathology, with murine modeling pointing to a role for the ERVK envelope (env) gene in disease processes. Here, we describe a novel viral protein cryptically encoded within the ERVK env transcript, which resembles two distinct cysteine-rich neurotoxic proteins: conotoxin proteins found in marine snails and the Human Immunodeficiency Virus (HIV) Tat protein. Consistent with Nuclear factor-kappa B (NF-κB)-induced retrotransposon expression, the ERVK conotoxin-like protein (CTXLP) is induced by inflammatory signaling. CTXLP is found in the nucleus, impacting innate immune gene expression and NF-κB p65 activity. Using human autopsy specimens from patients with ALS, we further showcase CTXLP expression in degenerating motor cortex and spinal cord tissues, concomitant with inflammation linked pathways, including enhancement of necroptosis marker mixed lineage kinase domain-like (MLKL) protein and oligodendrocyte maturation/myelination inhibitor Nogo-A. These findings identify CTXLP as a novel ERVK protein product, which may act as an effector in ALS neuropathology.
Collapse
|
25
|
Differences in splicing defects between the grey and white matter in myotonic dystrophy type 1 patients. PLoS One 2020; 15:e0224912. [PMID: 32407311 PMCID: PMC7224547 DOI: 10.1371/journal.pone.0224912] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multi-system disorder caused by CTG repeats in the myotonic dystrophy protein kinase (DMPK) gene. This leads to the sequestration of splicing factors such as muscleblind-like 1/2 (MBNL1/2) and aberrant splicing in the central nervous system. We investigated the splicing patterns of MBNL1/2 and genes controlled by MBNL2 in several regions of the brain and between the grey matter (GM) and white matter (WM) in DM1 patients using RT-PCR. Compared with amyotrophic lateral sclerosis (ALS, as disease controls), the percentage of spliced-in parameter (PSI) for most of the examined exons were significantly altered in most of the brain regions of DM1 patients, except for the cerebellum. The splicing of many genes was differently regulated between the GM and WM in both DM1 and ALS. In 7 out of the 15 examined splicing events, the level of PSI change between DM1 and ALS was significantly higher in the GM than in the WM. The differences in alternative splicing between the GM and WM may be related to the effect of DM1 on the WM of the brain.
Collapse
|
26
|
Chen QF, Zhang XH, Huang NX, Chen HJ. Identification of Amyotrophic Lateral Sclerosis Based on Diffusion Tensor Imaging and Support Vector Machine. Front Neurol 2020; 11:275. [PMID: 32411072 PMCID: PMC7198809 DOI: 10.3389/fneur.2020.00275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/24/2020] [Indexed: 11/13/2022] Open
Abstract
Objectives: White matter (WM) impairments involving both motor and extra-motor areas have been well-documented in amyotrophic lateral sclerosis (ALS). This study tested the potential of diffusion measurements in WM for identifying ALS based on support vector machine (SVM). Methods: Voxel-wise fractional anisotropy (FA) values of diffusion tensor images (DTI) were extracted from 22 ALS patients and 26 healthy controls and served as discrimination features. The revised ALS Functional Rating Scale (ALSFRS-R) was employed to assess ALS severity. Feature ranking and selection were based on Fisher scores. A linear kernel SVM algorithm was applied to build the classification model, from which the classification performance was evaluated. To promote classifier generalization ability, a leave-one-out cross-validation (LOOCV) method was adopted. Results: By using the 2,400~3,400 ranked features as optimal features, the highest classification accuracy of 83.33% (sensitivity = 77.27% and specificity = 88.46%, P = 0.0001) was achieved, with an area under receiver operating characteristic curve of 0.862. The predicted function value was positively correlated with patient ALSFRS-R scores (r = 0.493, P = 0.020). In the optimized SVM model, FA values from several regions mostly contributed to classification, primarily involving the corticospinal tract pathway, postcentral gyrus, and frontal and parietal areas. Conclusions: Our results suggest the feasibility of ALS diagnosis based on SVM analysis and diffusion measurements of WM. Additional investigations using a larger cohort is recommended in order to validate the results of this study.
Collapse
Affiliation(s)
- Qiu-Feng Chen
- College of Computer and Information Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiao-Hong Zhang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Nao-Xin Huang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hua-Jun Chen
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
27
|
Fournier CN, Houser M, Tansey MG, Glass JD, Hertzberg VS. The gut microbiome and neuroinflammation in amyotrophic lateral sclerosis? Emerging clinical evidence. Neurobiol Dis 2020; 135:104300. [PMID: 30321601 DOI: 10.1016/j.nbd.2018.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/07/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Christina N Fournier
- Department of Neurology, Emory University, Department of Veteran Affairs Medical Center, Atlanta, GA, United States.
| | - Madelyn Houser
- Department of Physiology, Emory University, United States.
| | - Malú G Tansey
- Department of Physiology, Emory University, United States.
| | | | | |
Collapse
|
28
|
Yuan J, Amin P, Ofengeim D. Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat Rev Neurosci 2019; 20:19-33. [PMID: 30467385 DOI: 10.1038/s41583-018-0093-1] [Citation(s) in RCA: 619] [Impact Index Per Article: 103.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Apoptosis is crucial for the normal development of the nervous system, whereas neurons in the adult CNS are relatively resistant to this form of cell death. However, under pathological conditions, upregulation of death receptor family ligands, such as tumour necrosis factor (TNF), can sensitize cells in the CNS to apoptosis and a form of regulated necrotic cell death known as necroptosis that is mediated by receptor-interacting protein kinase 1 (RIPK1), RIPK3 and mixed lineage kinase domain-like protein (MLKL). Necroptosis promotes further cell death and neuroinflammation in the pathogenesis of several neurodegenerative diseases, including multiple sclerosis, amyotrophic lateral sclerosis, Parkinson disease and Alzheimer disease. In this Review, we outline the evidence implicating necroptosis in these neurological diseases and suggest that targeting RIPK1 might help to inhibit multiple cell death pathways and ameliorate neuroinflammation.
Collapse
Affiliation(s)
- Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| | - Palak Amin
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
29
|
Neurofilament Immunohistochemistry Followed by Luxol Fast Blue, for Staining Axons and Myelin in the Same Paraffin Section of Spinal Cord. Appl Immunohistochem Mol Morphol 2019; 28:562-565. [PMID: 31652146 DOI: 10.1097/pai.0000000000000814] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Many disorders of the central nervous system are characterized by both axonal pathology and demyelination. In assessing this concurrent pathology, techniques for staining axons or myelin are frequently used separately. Here we report the development of a combined immunohistochemical and tinctorial staining technique in which we have modified the Luxol fast blue myelin stain to be used in conjunction with a diaminobenzidine-based immunohistochemical stain for high molecular weight neurofilament (SMI-31). This modification of staining will have utility in experimental neuropathology laboratories investigating demyelination and axonal damage in human tissue and animal models.
Collapse
|
30
|
Du XQ, Zou TX, Huang NX, Zou ZY, Xue YJ, Chen HJ. Brain white matter abnormalities and correlation with severity in amyotrophic lateral sclerosis: An atlas-based diffusion tensor imaging study. J Neurol Sci 2019; 405:116438. [PMID: 31484082 DOI: 10.1016/j.jns.2019.116438] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/23/2019] [Accepted: 08/28/2019] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To assess microstructural alterations in white matter (WM) in amyotrophic lateral sclerosis (ALS) using diffusion tensor imaging (DTI). METHODS DTI data were collected from 34 subjects (18 patients with ALS and 16 healthy controls). The atlas-based region of interest (ROI) analysis was conducted to assess WM microstructure in ALS by combining intra-voxel metrics, which included fractional anisotropy (FA) and mean diffusivity (MD), and an inter-voxel metric, i.e., local diffusion homogeneity (LDH). Correlation analysis of diffusion values and clinical factors was also performed. RESULTS ALS group showed a significant FA reduction in bilateral corticospinal tract (CST) as well as right uncinate fasciculus (RUF). The areas with higher MD were situated in right corticospinal tract (RCST), left cingulum hippocampus (LCH), RUF, and right superior longitudinal fasciculus (RSLF). Additionally, ALS patients showed decreased LDH in bilateral anterior thalamic radiation (ATR), bilateral CST and left inferior frontal-occipital fasciculus (LIFOF). Significant correlations were observed between ALSFRS-R (revised ALS Functional Rating Scale) scores or progression rate and FA in bilateral CST, as well as between disease duration and LDH in right CST. Receiver operating characteristic (ROC) analysis revealed the feasibility of employing diffusion metrics along the CST to distinguish two groups (AUC = 0.792-0.868, p < .005 for all). CONCLUSIONS WM microstructural alteration is a common pathology in ALS, which can be detected by both intra- and inter-voxel diffusion metrics. The extent of abnormalities in several WM tracts such as ATR and LIFOF may be better assessed through the inter-voxel diffusion measurement.
Collapse
Affiliation(s)
- Xiao-Qiang Du
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Tian-Xiu Zou
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Nao-Xin Huang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhang-Yu Zou
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| | - Yun-Jing Xue
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| | - Hua-Jun Chen
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| |
Collapse
|
31
|
Qiu T, Zhang Y, Tang X, Liu X, Wang Y, Zhou C, Luo C, Zhang J. Precentral degeneration and cerebellar compensation in amyotrophic lateral sclerosis: A multimodal MRI analysis. Hum Brain Mapp 2019; 40:3464-3474. [PMID: 31020731 PMCID: PMC6865414 DOI: 10.1002/hbm.24609] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/28/2019] [Accepted: 04/16/2019] [Indexed: 12/27/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive and intractable neurodegenerative disease of human motor system characterized by progressive muscular weakness and atrophy. A considerable body of research has demonstrated significant structural and functional abnormalities of the primary motor cortex in patients with ALS. In contrast, much less attention has been paid to the abnormalities of cerebellum in this disease. Using multimodal magnetic resonance imagining data of 60 patients with ALS and 60 healthy controls, we examined changes in gray matter volume (GMV), white matter (WM) fractional anisotropy (FA), and functional connectivity (FC) in patients with ALS. Compared with healthy controls, patients with ALS showed decreased GMV in the left precentral gyrus and increased GMV in bilateral cerebellum, decreased FA in the left corticospinal tract and body of corpus callosum, and decreased FC in multiple brain regions, involving bilateral postcentral gyrus, precentral gyrus and cerebellum anterior lobe, among others. Meanwhile, we found significant intermodal correlations among GMV of left precentral gyrus, FA of altered WM tracts, and FC of left precentral gyrus, and that WM microstructural alterations seem to play important roles in mediating the relationship between GMV and FC of the precentral gyrus, as well as the relationship between GMVs of the precentral gyrus and cerebellum. These findings provided evidence for the precentral degeneration and cerebellar compensation in ALS, and the involvement of WM alterations in mediating the relationship between pathologies of the primary motor cortex and cerebellum, which may contribute to a better understanding of the pathophysiology of ALS.
Collapse
Affiliation(s)
- Ting Qiu
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduPeople's Republic of China
| | - Yuanchao Zhang
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduPeople's Republic of China
| | - Xie Tang
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduPeople's Republic of China
| | - Xiaoping Liu
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduPeople's Republic of China
| | - Yue Wang
- Key Laboratory for NeuroInformation of Ministry of Education, School of Life Science and TechnologyUniversity of Electronic Science and Technology of ChinaChengduPeople's Republic of China
| | - Chaoyang Zhou
- Department of RadiologySouthwest Hospital, Third Military Medical UniversityChongqingPeople's Republic of China
| | - Chunxia Luo
- Department of NeurologySouthwest Hospital, Third Military Medical UniversityChongqingPeople's Republic of China
| | - Jiuquan Zhang
- Department of RadiologyChongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer HospitalChongqingPeople's Republic of China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University)Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer HospitalChongqingPeople's Republic of China
| |
Collapse
|
32
|
Are Astrocytes the Predominant Cell Type for Activation of Nrf2 in Aging and Neurodegeneration? Antioxidants (Basel) 2017; 6:antiox6030065. [PMID: 28820437 PMCID: PMC5618093 DOI: 10.3390/antiox6030065] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 12/29/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that regulates hundreds of antioxidant genes, and is activated in response to oxidative stress. Given that many neurodegenerative diseases including Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, Huntington’s disease and multiple sclerosis are characterised by oxidative stress, Nrf2 is commonly activated in these diseases. Evidence demonstrates that Nrf2 activity is repressed in neurons in vitro, and only cultured astrocytes respond strongly to Nrf2 inducers, leading to the interpretation that Nrf2 signalling is largely restricted to astrocytes. However, Nrf2 activity can be observed in neurons in post-mortem brain tissue and animal models of disease. Thus this interpretation may be false, and a detailed analysis of the cell type expression of Nrf2 in neurodegenerative diseases is required. This review describes the evidence for Nrf2 activation in each cell type in prominent neurodegenerative diseases and normal aging in human brain and animal models of neurodegeneration, the response to pharmacological and genetic modulation of Nrf2, and clinical trials involving Nrf2-modifying drugs.
Collapse
|